WO2004081050A1 - Anticorps contre un antigene specifique aux tumeurs - Google Patents

Anticorps contre un antigene specifique aux tumeurs Download PDF

Info

Publication number
WO2004081050A1
WO2004081050A1 PCT/JP2004/003048 JP2004003048W WO2004081050A1 WO 2004081050 A1 WO2004081050 A1 WO 2004081050A1 JP 2004003048 W JP2004003048 W JP 2004003048W WO 2004081050 A1 WO2004081050 A1 WO 2004081050A1
Authority
WO
WIPO (PCT)
Prior art keywords
antibody
human
cells
cancer
protein
Prior art date
Application number
PCT/JP2004/003048
Other languages
English (en)
Japanese (ja)
Inventor
Kimihisa Ichikawa
Shu Takahashi
Toshinori Agatsuma
Keisuke Fukuchi
Takehiro Hirai
Original Assignee
Sankyo Company Limited
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority to AU2004220182A priority Critical patent/AU2004220182B2/en
Priority to EP04718767A priority patent/EP1602669B1/fr
Application filed by Sankyo Company Limited filed Critical Sankyo Company Limited
Priority to US10/548,688 priority patent/US7855056B2/en
Priority to BRPI0408238-9A priority patent/BRPI0408238A/pt
Priority to MXPA05009715A priority patent/MXPA05009715A/es
Priority to DE602004021567T priority patent/DE602004021567D1/de
Priority to CA002518787A priority patent/CA2518787A1/fr
Priority to AT04718767T priority patent/ATE434044T1/de
Priority to NZ542220A priority patent/NZ542220A/en
Priority to DK04718767T priority patent/DK1602669T3/da
Publication of WO2004081050A1 publication Critical patent/WO2004081050A1/fr
Priority to US11/223,812 priority patent/US7361340B2/en
Priority to NO20054631A priority patent/NO20054631L/no
Priority to HK06100346.5A priority patent/HK1078593A1/xx
Priority to US11/345,651 priority patent/US20070025996A1/en
Priority to US11/872,479 priority patent/US7741447B2/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/32Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against translation products of oncogenes
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/42Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against immunoglobulins
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • C12Q1/6886Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material for cancer
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • C07K2317/732Antibody-dependent cellular cytotoxicity [ADCC]
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/136Screening for pharmacological compounds
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/158Expression markers

Definitions

  • the present invention relates to an antibody that can be used for cancer treatment, a pharmaceutical composition for treating cancer, which comprises the body as an active ingredient, a method for detecting cancer, and a kit for detecting cancer.
  • tumor cells express a unique antigen protein (hereinafter, sometimes referred to as a tumor-associated antigen) depending on the cell type. Therefore, attempts are being made to develop new therapeutic methods for tumors by using this tumor-associated antigen as a target.
  • Monoclonal monocytes that show a specific antigen-antibody response to tumor-associated bovine cells have various biological immune reactions (antibody-dependent cellular cytotoxicity (ADCC), complement-dependent cytotoxicity ( It is known that it induces cancer cells by inducing CDC), etc., and induces cell death.
  • ADCC antibody-dependent cellular cytotoxicity
  • CDC complement-dependent cytotoxicity
  • Monoclonal K-forms useful for treating tumors have been developed.
  • monoclonal antibodies useful for the treatment of epilepsy are limited to several tumor types, such as metastatic breast cancer, acute myeloid leukemia, refractory chronic lymphoma, non-Hodgkin's lymphoma, and multiple myeloma.
  • tumor types such as metastatic breast cancer, acute myeloid leukemia, refractory chronic lymphoma, non-Hodgkin's lymphoma, and multiple myeloma.
  • monoclonal antibody that can be used for treating other tumors.
  • Human oculospanin is a protein obtained as an Expressed Sequence Tag (EST) clone from genes expressed in the retinal pigment epithelium and ocular choroid (Molecular Vision, (2002) 8, 25-220).
  • the human oculospanin gene has an open-reading frame of 1068 bp, and the human oculospanin has a length of amino acid from 355 amino acids.
  • the molecular weight estimated from the DNA sequence is 36.4 kDa, the relationship between human oculospanin and tumor has not been clarified. Disclosure of the invention
  • the present invention provides a method for detecting a cancer by detecting a gene that is specifically expressed in a cancer, and detecting the expression of the gene, a kit for detecting a cancer used in the detection method, and a method for specifically expressing an expression product of the gene.
  • An object of the present invention is to provide an antibody that binds and has cytotoxic activity, and a pharmaceutical composition for treating cancer comprising the antibody as an active ingredient.
  • the present inventors searched for a gene that is specifically expressed in human cancer tissues, and the expression level of the human oculospanin gene, whose function was unknown, has been significantly increased in melanocytic cells. Accordingly, the present invention provides a method for detecting cancer using the gene, a kit for detecting cancer, and a pharmaceutical composition for treating cancer, which contains an anti-human oculospanin antibody, thereby completing the present invention.
  • cytotoxic activity is apoptosis induction, (1) or
  • a method for detecting cancer comprising the following steps 1) to 4):
  • a method for detecting cancer comprising the following steps 1) to 3):
  • step 2) measuring the expression level of at least one of the proteins described in step 1) above in a sample collected from a normal person;
  • the method for measuring the expression level of a polynucleotide is characterized in that it is a Northern plot method, a dot plot method, a slot blot method, RT-PCR, a liponuclease-protected assay or a run-on assay. , (12), (14) and (15);
  • the method for measuring the expression level of a polynucleotide is characterized in that a gene chip or an array made of DNA comprising a partial sequence of each DNA of a complementary DNA group or a DNA group derived from a specimen is used. , (12), (14) and (15);
  • a kit for detecting a cancer containing at least one or more selected from the group consisting of the following 1) to 3):
  • Kit for detecting cancer containing at least one of the following 1) and 2): 1) Protein consisting of the amino acid sequence shown in SEQ ID NO: 2 in Sequence Listing and SEQ ID NO in Sequence Listing Specifically binds to a protein consisting of the amino acid sequence shown in 4, A body for detecting the protein;
  • a pharmaceutical composition for treating cancer comprising at least one of the antibodies according to (1) to (11).
  • a pharmaceutical composition for treating cancer comprising an oligonucleotide having a nucleotide sequence represented by SEQ ID NO: 1 in the Sequence Listing or a nucleotide sequence complementary to a partial distribution sequence of the sequence,
  • the upper diagram of FIG. 1 is a graph showing the expression of the human oculospanin gene in various cell lines.
  • the lower diagram of FIG. 1 is a graph showing the expression levels of the human oculospanin gene in healthy human skin samples and melanophora samples.
  • the upper diagram of FIG. 2 is a graph showing the expression level of the human oculospanin gene in a healthy human skin sample and a skin tissue-derived melanoma sample.
  • the lower diagram of FIG. 2 is a graph showing the expression level of the human oculospanin gene in a healthy human skin sample and a lymphoma node-derived melanoma sample.
  • FIG. 3 is a graph showing the expression level of the human occulospanin gene in a sample derived from a healthy human lymph node and a melanosample derived from a lymph node tissue.
  • FIG. 4 is a view showing expression of a human occulospanin gene product in NIH3T3 cells.
  • Figure 5 shows the results of anti-oculospanis using cells expressing human oculospanins.
  • 4 is a graph showing the antibody-dependent cytotoxic activity of the antibody.
  • a compound having a therapeutic effect on cancer refers to a compound having an activity of suppressing cancer growth and an activity of reducing cancer.
  • cancer and “tumor” are used interchangeably.
  • the term “gene” is intended to include not only DNA but also its mRNA, cDNA and its cRNA. Therefore, the term “human oculospanin gene J” in the present invention includes DNA, mRNA, cDNA and cDNA of human oculospanin.
  • polynucleotide refers to a nucleic acid. They are used interchangeably and include DNA, RNA, probes, oligonucleotides, and primers.
  • RNA fraction refers to a fraction containing RNA.
  • cells includes cells in an animal individual and cultured cells.
  • cell carcinogenesis means that cells exhibit abnormal growth, such as losing sensitivity to the contact inhibition phenomenon or exhibiting anchorage-independent growth. A cell exhibiting such abnormal growth is referred to as “cancer cell J.
  • a protein having a function equivalent to the oncogenic activity of a cell having human oculospanin is also referred to as a human oculospanin.
  • oncogene (Oncoge) in the present invention includes oncogenes and proto-oncogenes (Proto-Oncogene) in addition to oncogenes.
  • cytotoxicity refers to causing pathological changes in cells in any way, not only direct trauma but also DNA cleavage, base dimer formation, and chromosomal damage. This refers to any structural or functional damage to any cell, such as cleavage, damage to cell division machinery, or reduced activity of various enzymes. “Cytotoxic activity” in the present invention means causing the above-mentioned cell injury.
  • hybridizing under stringent conditions means that hybridization is performed at 68 ° C. in a commercially available hybridization solution ExpressHyb Hybridization Solution (manufactured by Clontech), or DNA is synthesized. Hybridization at 68 ° C in the presence of 0.7-1.0 M NaC1 using a fixed filter After performing the daidze reaction, use 0.1- to 2-fold concentration SSC solution (1x concentration SSC consists of 150 mM NaCl and 15 mM sodium citrate), and Hybridization under conditions that can be identified by washing with C or equivalent conditions.
  • the expression level of the human occulospanin gene in various human cell lines was significantly higher in melanocytes than in other tissues, and moreover in melanomas than in normal melanocytes. It has been found by the present inventors that the expression level is significantly increased. For example, comparing the expression levels of human oculospanin among melanocytes, lymphoblasts, glial cells, and epithelial cells, the expression levels are significantly higher in melanocytes, and normal skin cells and melanoma cells. Comparing the expression levels of human oculospanin in the above, the present inventors found that the expression level was remarkably high in melanomas.
  • human oculospanin is involved in canceration of cells and / or proliferation of cancer cells. That is, by measuring the expression level of human occulospanin in each cell and / or each tissue, the state of canceration and / or proliferation of cancer cells caused by overexpression of human occulospanin can be determined. Can be determined.
  • Such cancers include, for example, skin cancers, in particular, melanoma. If the expression of human occulospanin is significantly greater than that of other tissues, other cancers may be used. It can also be applied to cancer.
  • the nucleotide sequence of the open reading frame (0RF) of the human oculospanin gene is shown in SEQ ID NO: 1 in the sequence listing, and its amino acid sequence is shown in SEQ ID NO: 2 in the sequence listing.
  • OCSP Homo sapiens oculospanin
  • mRNA accession number: NM_ 0 3 1 9 4 5
  • GenBank accession number: NM_ 0 3 1 9 4 5
  • the nucleotide sequence of the cDNA used is also shown in SEQ ID NO: 3 in the sequence listing, and 0RF is shown in nucleotide numbers 65 to 112 of SEQ ID NO: 3.
  • Hit Oculus registered in GenBank An amino acid sequence identical to the amino acid sequence of panin is shown in SEQ ID NO: 4 in the sequence listing.
  • amino acid sequence of human oculospanin one or several amino acids are substituted, deleted or added, and the protein having the same biological activity as human oculospanin is also a protein of human oculospanin. include. .
  • human oculospanin is highly expressed in cancer cells, particularly in melanoma, it is considered to be involved in canceration of cells, particularly skin cells, and / or proliferation of cancer cells.
  • the ⁇ sample '' refers to blood, body fluid, prostate, testis, penis, bladder, kidney, oral cavity, pharynx, lips, tongue, gingiva, nasopharynx, esophagus, stomach, Tissues or excretions such as small intestine, large intestine, colon, liver, gallbladder, Teng, nose, lung, bone, soft tissue, skin, breast, uterus, ovary, brain, thyroid, lymph node, muscle, adipose tissue, etc.
  • the skin or the lymph node is more preferable in the present invention.
  • Method for detecting cancer using the expression level of human occulospanin gene The method for detecting cancer using the expression level of the human occulospanin gene is specifically described in the following steps 1) to 4) It is a method including.
  • Step 1) Extracting the total RNA fraction from the sample collected from the subject;
  • human-derived tissue obtained by a method appropriate to the appropriate experimental ethical standards Is directly dissolved in a solvent for RNA extraction (for example, one containing a component that inactivates liponuclease, such as phenol).
  • the cells were collected by a method such as gently squeezing with a scraper or gently extracting the cells from the tissue with a protease such as trypsin so as not to destroy the cells in the tissue. Then, immediately proceed to the RNA extraction step. .
  • RNA extraction methods include guanidine thiocyanate 'cesium chloride ultracentrifugation method, guanidine thiocyanate-hot phenol method, guanidine hydrochloride method, and acid guanidine thiocyanate-phenol-clonal form method (Chomczynski, P. an d Sacc iN, Anal. Biochem. (1987), 162, 156-159), etc., but the acid guanidine thiocyanate / phenol / chloroform method is preferred.
  • reagents for RNA extraction for example, ISOGEN (manufactured by Futtsubon Gene Co., Ltd.), TRIZOL reagents (manufactured by Gibco 'PRL)) and the like can also be used according to the protocol attached to the reagents.
  • ISOGEN manufactured by Futtsubon Gene Co., Ltd.
  • TRIZOL reagents manufactured by Gibco 'PRL
  • the obtained total RNA fraction is preferably further purified and used only for mRNA if necessary.
  • the purification method is not particularly limited.
  • mRNA is adsorbed to a biotinylated oligo (dT) probe, and streptavidin-immobilized paramagnetic particles are used by utilizing the binding between biotin and streptavidin.
  • the mRNA can be purified by eluting the mRNA.
  • a method of adsorbing mRNA onto an oligo (dT) cellulose column and then eluting the mRNA and purifying it may be employed.
  • the step of purifying these mRNAs is not essential for the method of the present invention, and the total RNA fraction should be used in the subsequent steps as long as the expression of the target polynucleotide can be detected. You can also. '
  • a normal person means a person who does not have cancer. Whether a person is normal or not can be determined by measuring the concentration of human oculospanin and determining whether or not the value falls within a numerical range determined in advance as a normal person. The correlation between the expression level of the gene and the degree of cancer formation in normal subjects is checked in advance, and the subject is a normal subject by measuring the expression level of human oculospanin in a sample collected from the subject. Or not. Preparation of the total RNA fraction from normal subjects should be performed in the same manner as in step 1) above. Can be.
  • Step 3) A step of measuring the expression level of the human occulospanin gene in the total RNA fraction described in step 1) above and the total RNA fraction described in step 2) above: here, human occulospanin
  • the expression level of the gene is determined by the polynucleotide comprising the nucleotide sequence shown in SEQ ID NO: 1 in the Sequence Listing, or the polynucleotide comprising the nucleotide sequence complementary to the nucleotide sequence shown in SEQ ID NO: 1 in the Sequence Listing and the string. This is indicated by the expression level of a polynucleotide that hybridizes under various conditions. '
  • Examples of the immobilized sample include the following.
  • a gene chip on which an antisense oligonucleotide synthesized on the basis of an EST (expressedseqsecncetag) sequence or mRNA sequence on a database is immobilized.
  • a gene chip is a gene chip manufactured by Affymetrix (A fymetrix) (Lip shut z,. J. et al., Nature Genet. (1999) 21, suppl iment, 20-24). ) Can be used, but the present invention is not limited to this, and it may be produced based on a known method.
  • human-derived mRNA is preferable.
  • human U95 set or U133 set manufactured by Affymetrix can be used.
  • the invention is not limited thereto, and for example, those derived from closely related animals can be used.
  • cDNA or RT-PCR product prepared from human, total RNA, or total RNA obtained from a specific tissue. It was cloned by performing a reverse transcriptase reaction or PCR using a primer prepared based on the sequence information of the EST database and other information.
  • This cDNA and RT-PCR product Differential expression of total RNA between humans with and without tumors was determined by the subtraction method (Diatchenki, L, et al, Proc. Natl. Acad. Sci. USA, (1996) 93, 6025-6030) and the differential display method (Liang, P., et al Nucleic Acids Res., (1992) 23, 3685-3690). Good.
  • the labeled probe use not a specific mRNA clone but a label of all expressed mRNAs.
  • unpurified mRNA may be used as a starting material for preparing the cap, it is preferable to use poly (A) + RNA purified by the method described above.
  • poly (A) + RNA purified a method for preparing a labeled probe and a detection and analysis method using various types of immobilized samples will be described.
  • a biotin-labeled cRNA probe is prepared according to the protocol (Affimetrix Expression Analysis Technical Manual) attached to the Affimetrix GeneChip. Then, according to the protocol (Expression Analysis Technology Manual) attached to the gene chip manufactured by Affimetrics, hybridization was performed using an analysis device (GeneChip Fluidics Station 400) manufactured by Affymetrix. Perform an analysis and analysis to detect and analyze luminescence by avidin.
  • cDNA When preparing cDNA from poly (A) + RNA in reverse transcriptase reaction, it is necessary to label cDNA so that cDNA can be detected.When labeling with fluorescent dye Then, dDNA is labeled with a fluorescent dye (eg, Cy3, Cy5, etc.), and the cDNA is fluorescently labeled. At this time, if poly (A) + RNA derived from melanoma cells and poly (A) + RNA derived from control cells are labeled with different dyes, both will be mixed during the subsequent hybridization. Can be used.
  • a fluorescent dye eg, Cy3, Cy5, etc.
  • the array to be used is not limited to a commercially available array, and may be a home-made array or a specially prepared array.
  • a labeled isotope is prepared by adding a radioisotope (for example, d-CTP, etc.), and hybridization is performed by a conventional method.
  • a radioisotope for example, d-CTP, etc.
  • hybridization is performed by a conventional method.
  • the method according to any one of the above (a) to (c) hybridizes a probe derived from each human tissue to the solid-phased sample of the same lot. At this time, the conditions other than the hybridization used except for the hybridization are the same.
  • a fluorescently labeled probe if each probe is labeled with a different fluorescent dye, the mixture of both probes can be hybridized to one solid-phased sample at a time, and the fluorescence intensity can be read ( Brown, P.0. Et al. Nature Genet., (1999) 21, supplement, p.33-37).
  • Step 4) The difference in the expression level of the gene measured in the above step 3) between the total RNA fraction derived from the above step 1) and the total RNA fraction derived from the above step 2) is analyzed, and the above step 1) is analyzed. 4. The step of detecting cancer in a subject according to the above.
  • a cancer in which the expression level of human oculospanin is significantly increased is cancer, especially skin cancer. Further, it can be determined that there is a high possibility that melanoma is present, and cancer can be detected.
  • the expression level is significantly increased, for example, when analysis is performed using Affimetrix's gene chip and Affimetrix's microarray Suite Ver. This means that the averagedifference value of the cell-derived gene is significantly increased as compared to normal melanocytes.
  • the concentration of the human occulospanin gene is measured, and it is analyzed whether or not the value falls within the numerical range determined in advance as a normal value.
  • a cancer can be detected by determining that it has cancer, or a sample collected from a subject by previously examining the correlation between the expression level of the human occulospanin gene and the degree of cancer formation in normal humans. By measuring the expression level of the human occulospanin gene in the above, it can be determined whether or not the subject is a normal person.
  • a method for detecting cancer using the expression level of human occulospanin is, specifically, a method including the following steps 1) to 3).
  • sample After removing insoluble substances by performing high-speed centrifugation as necessary, prepare the sample as a sample for ELISA / RIA or a sample for Westin blot as follows.
  • the recovered skin or lymph node may be used as it is, or may be appropriately diluted with a buffer.
  • Western blot (electrophoresis) samples for example, use the extract of skin or lymph node as it is, or dilute appropriately with buffer, and use SDS-polyacrylamide electrophoresis 21-mercator. Mix with a sample buffer containing ethanol (eg, from Sigma).
  • a dot / slot plot for example, the collected skin or lymph node extract itself or appropriately diluted with a buffer solution is directly adsorbed to the membrane using a plotting device.
  • the sample is precipitated by immunoprecipitation, a method using ligand binding, etc., and detected without immobilization.
  • the sample to be detected can be immobilized as it is.
  • a nitrocellulose membrane e.g., manufactured by Bio-Rad Co., Ltd.
  • a nylon membrane e.g., etc.
  • Hybond-I ECL (Amersha-Pharmacia) etc.), Cotton Membrane (for example, Protosorbent Filter-1 (Bio-Rad) etc.), or Polyvinylidene Difluoride (PVDF) membrane (for example, Bio-Rad) And the like).
  • Cotton Membrane for example, Protosorbent Filter-1 (Bio-Rad) etc.
  • PVDF Polyvinylidene Difluoride
  • a 96-well plate for example, Imnoplate / Maxisorp (produced by Nunc) is used to prepare a sample or a dilution thereof (for example, 0.05).
  • % Phosphate buffered saline containing sodium azide diluted in PBS
  • the protein is adsorbed on the inner bottom surface of the well to be immobilized by leaving it overnight or at 37 for 1 to 3 hours.
  • Antibodies to human oculospanin can be obtained from the amino acid sequence of human oculospanin or human oculospanin using a conventional method (see, for example, Shinsei Kagaku Kenkyusho 1, Protein 1, p. 389-397, 1992). It can be obtained by immunizing an animal with any selected polypeptide and collecting and purifying antibodies produced in the living body. Further, according to a known method (for example, Kohler and Milstein, Nature 256, 495-497, 1975, Kennet, R. ed., Monoclonal Ant ibody p.
  • a monoclonal antibody can also be obtained by establishing a hybridoma by fusing an antibody-producing cell that produces an antibody against oculospanin with a myeloma cell.
  • human oculospanin as an antigen can be obtained by producing a human oculospanin gene in a host cell by genetic manipulation. Specifically, a vector capable of expressing a human oculospanin gene can be prepared, introduced into a host cell to express the gene, and the expressed human oculospanin can be purified.
  • the expression level of human oculospanin is represented by expression a of a protein comprising an amino acid sequence shown in SEQ ID NO: 2 in the sequence listing.
  • the expression level can be measured using the above-mentioned anti-human occulospanin antibody by a known method such as the western blot method / dot / slot blot method.
  • Step 2) Step of measuring the expression level of the protein described in 1) above in a sample collected from a normal person:
  • the measurement of the expression level of human touloculospanin in a sample collected from a normal person can be performed in the same manner as in the above step 1).
  • Step 3) a step of analyzing the difference between the expression level of the protein measured in the above step 1) and the expression S of the protein measured in the above step 2) to detect cancer in the subject.
  • the concentration of human oculospanin is measured, and it is analyzed whether or not the concentration falls within the numerical range determined in advance as a normal person value.
  • Cancer can be detected by determining that it has cancer, or by examining in advance the correlation between the expression level of human * oculospanin and the degree of cancer formation in normal humans, it is possible to detect human cancer in a sample collected from a subject. • It is also possible to determine whether or not the subject is a normal person by measuring the expression level of toulospanin.
  • a human cDNA library derived from a cell expressing human ococulospanin is prepared according to a known method such as colony hybridization method, etc. Obtain full-length cDNA. This full-length cDNA is introduced into a mouse or a human cell to be highly expressed, and it is examined whether or not the cell is affected.
  • a method of incorporating the obtained full-length cDNA into a virus vector and administering it to an animal can be mentioned.
  • a virus vector for example, cDNA is incorporated into a DNA virus such as a retrovirus, an adenovirus, an adeno-associated virus, a herpes virus, a vaccinia virus, a box virus, a polio virus, or an RNA virus.
  • the method introduced in is mentioned. Among them, methods using retrovirus, adenovirus, adeno-associated virus, and vaccinia virus are preferable.
  • Non-viral gene transfer methods include a method of directly expressing expression plasmid into muscle (DNA vaccine method), Ribosome method, Lipofection method, microinjection method, calcium phosphate method, electoral poration method, etc. Among them, the DNA vaccine method and the ribosome method are preferred.
  • full-length cDNA is introduced into muscle cells, hepatocytes, adipocytes, or muscle cells, hepatocytes, adipocytes, skin cells, etc. derived from human, mouse, rat, etc. Highly expressed, the function of each target cell, specifically, the function of regulating glycolipid metabolism such as production and uptake of glycolipids or accumulation of glycogen, or what effect on cell morphology You can consider whether it will appear. Conversely, an antisense nucleic acid against all R R ⁇ of the gene to be tested can be introduced into cells, and the effect on the function and morphology of each target cell can be examined.
  • the cDNA When introducing full-length cDNA into an animal or a cell, the cDNA is incorporated into a vector containing an appropriate promoter and a sequence involved in expression of the gene, and the vector is used to transform a host cell.
  • a vertebrate cell expression promoter one having a promoter located upstream of a gene to be normally expressed, an RNA splice site, a polyadenylation site, and a transcription termination sequence can be used. May have a replication origin if necessary.
  • An example of such an expression vector is pSV2dhfr (Subramani, S. et al., Mol. Cell. Biol., (1981) 1, .854-864) which has an initial promoter of SV40.
  • Retrovirus vectors pLNCX, pLNSX, pLXIN, pSIR manufactured by Clontech
  • cosmid vector-1 pAxCw manufactured by Takara Bio Inc.
  • the expression vector is prepared by the method of acetylaminoethyl (DEAE) -Axtran (Luthman, H. and Magnusson, G., Nucleic Acids Res. (1983) 11, .1295-1308), Calcium-DNA co-precipitation method (Graham, FL and van der Eb, AJ Virology, (1973) 52, p.456-457), and electric pulse perforation method (Neumann, E.
  • COS cells COS cells (Gluzman, Y. Cel 1 (1981) 23, p.175-182, ATCC: CRL-1650), which are monkey cells, and Chinese hamster ovary cells (1, .841-845).
  • CH ⁇ cell, AT CC: CCL-161) dihydrofolate reductase-deficient strain Urlaub, G. and Chasin, LA Pro c. Natl. Acad. Sci. USA (1980) 77, p.4126-4220
  • It can be incorporated into human fetal kidney-derived 293 cells (ATCC: CRL-15773) and the like, but is not limited thereto. Thus, a desired transformant can be obtained.
  • the target gene is highly expressed in normal animals by genetic manipulation. It is also possible to prepare transgenic animals in such a way as to study the effects on cell morphology. Conversely, in an animal having melanoma, a knockout animal in which the target gene has been disrupted can be prepared to determine the state of the cell.
  • the human oculospanin gene and / or the human oculospanin can be detected using a kit containing at least one selected from the group consisting of the following 1) to 5).
  • a continuous oligonucleotide primer having a length of 15 to 30 bases for specifically amplifying a polynucleotide consisting of the nucleotide sequence shown in SEQ ID NO: 1 in the sequence listing;
  • the primer described in 1) above is a commercially available primer design software (for example, Wisconsin) based on the nucleotide sequence of the human occulospanin gene (the nucleotide sequence shown in SEQ ID NO: 1 in the sequence listing). It can be easily designed and amplified by conventional methods, such as by using GCG package Ver. 10.2).
  • a primer for example, in order to amplify a polynucleotide consisting of the nucleotide sequence shown in SEQ ID NO: 1 in the Sequence Listing, an oligonucleotide consisting of the nucleotide sequence shown in SEQ ID NO: 5 in the Sequence Listing is used.
  • a combination of oligonucleotides consisting of the nucleotide sequence shown in SEQ ID NO: 6 in the column list can be used.
  • the probe described in 2) above specifically hybridizes to human oculospanin. It is a polynucleotide to be hybridized and has a length of 100 to 150 bases, preferably 300 to 600 bases.
  • These primers and probes may be labeled with an appropriate label (for example, an enzyme label, a radioactive label, a fluorescent label, etc.), or may be linked with a linker.
  • the solid-phased sample described in 3) above is prepared by immobilizing the probe described in 2) above on a solid phase such as a glass plate or nylon membrane.
  • the method for preparing such an immobilized sample has already been described in the section “(1) Cancer detection method using the expression level of human occulospanin gene” in the section “2. Cancer detection method”.
  • (1) Measurement method using solid-phased sample for example, a gene chip, a cDNA array, an oligo array, a membrane filter, and the like.
  • the kit of the present invention can further include a thermostable DNA polymerase, dNTP (a mixture of dATP, dCTP, dGTP, and dTTP) and a buffer.
  • dNTP a mixture of dATP, dCTP, dGTP, and dTTP
  • the heat-resistant DNA polymerase include Taq DNA polymerase, LAT aq DNA pollym erase (manufactured by Takara Shuzo), Tth DNA polymerase, and Pfu DNA polymerase.
  • the buffer is selected according to the DNA polymerase used, and Mg 2+ or the like is added as necessary.
  • the antibody described in 4) and 5) above is a method for detecting cancer using the expression level (3) of human oculospanin (protein expression S) in the above section “2. Method for detecting cancer”. And the method described in “5. Production of human occulopanin antibody”.
  • the antibody may be labeled with an appropriate label (for example, an enzyme
  • the kit of the present invention can be used for the detection of the human occulospanin gene and / or the human occlusion spanin, and is also used for determining the presence or absence of cancer and for screening for a substance that suppresses cancer growth. be able to.
  • a human oculopanin or a polypeptide comprising at least six consecutive partial amino acid sequences thereof is used as an antigen for producing a human oculopanin antibody.
  • Peptides and derivatives obtained by adding an arbitrary amino acid sequence or a carrier to them can be given.
  • Human occulospanin can be used by directly purifying it from human tumor tissue or tumor cells, and can also be obtained by synthesizing human 'oculopanin in vitro or by producing it in host cells by genetic engineering.
  • human 'oculopanin is inserted into a vector capable of expressing it, and then synthesized in a solution containing enzymes, substrates and energy substances required for transcription and translation, or other prokaryotic organisms.
  • the protein can be obtained by expressing human oculopanin by transforming a eukaryotic host cell.
  • the nucleotide sequence of the cDNA of human 'oculopanin is described in the literature (Graeme Wistow, Steven L. Bernstein, M.
  • cDNA of human oculospanin can be obtained by using a cDNA library expressing human oculospanin as a type I and using a primer that specifically amplifies human oculospanin cDNA. It can be obtained by the so-called PCR method that performs a chain reaction (hereinafter referred to as ⁇ pCRJ) [see Saiki, RK, et al. (1988) Science 239, 487-49].
  • Examples of the in vitro synthesis of polypeptides include, but are not limited to, a rapid translation system (RTS) manufactured by Kyushu Diagnostics.
  • RTS rapid translation system
  • MRNA is transcribed by T7 RNA polymerase, then translated by liposomes in E. coli lysate, and the desired polypeptide is synthesized in the reaction solution (Biochemica, 1, 20). -23 (2001), Biochemica, 2, 28-29 (2001).
  • Prokaryotic host cells include, for example, Escherichia coli and Bacillus subtilis.
  • the host cell is transformed with a plasmid vector containing regulatory elements and a replicon or origin of replication compatible with the host.
  • the vector is preferably a vector having a sequence capable of imparting phenotypic (phenotypic) selectivity to transformed cells.
  • K12 strain or the like is often used as Escherichia coli
  • pBR322 or pUC-based plasmid is generally used as a vector, but is not limited thereto, and various known strains and vectors may be used. Can be used.
  • tryptophan (trp) promoter In Escherichia coli, tryptophan (trp) promoter, lactose (lac) promoter, tryptophan lactose (tac) promoter, lipoprotein (lpp) promoter, polypeptide chain extension H tufB) promoter and the like, and any promoter can be used for production of a desired polypeptide.
  • Bacillus subtilis for example, 207-25 strain is preferable, and as a vector, pTU ⁇ 228 (Ohmura, K. et al. (1984) J. Biochem. 95, 87-93) is used.
  • pTU ⁇ 228 Ohmura, K. et al. (1984) J. Biochem. 95, 87-93
  • the present invention is not limited to this.
  • extracellular secretory expression is also possible.
  • Eukaryotic host cells include cells such as vertebrates, insects, and yeast.
  • vertebrate cells include COS cells, which are monkey cells (Gluzman, Y. (198 1) Cell 23, 175 -182, AT CCCRL— 1650), mouse fibroblast NIH 3T3 (AT CCN o. CRL—168 5) and Chinese hamster ovary cells (CHO cells, AT CCCCL—61).
  • COS cells which are monkey cells (Gluzman, Y. (198 1) Cell 23, 175 -182, AT CCCRL— 1650), mouse fibroblast NIH 3T3 (AT CCN o. CRL—168 5) and Chinese hamster ovary cells (CHO cells, AT CCCCL—61).
  • Strain deficient in dihydrofolate reductase Urlaub, G. and Chasin, LA (1980) Proc. Natl. Acad. Sci. USA 77, 4126-4220 is often used, but is not limited thereto. .
  • RNA splice site As vertebrate cell expression promoters, those having a promoter located upstream of the gene to be normally expressed, an RNA splice site, a polyadenylation site, and a transcription termination sequence can be used. It may also have a replication origin if necessary.
  • An example of such an expression vector is the first Zytomegalovirus. 3.1 (produced by Invitrogen) having an early promoter and pSV2dhfr. (Subramani, S. et al. (1981) Mol. Cell. Biol. 1) having an early promoter of SV40. , 854-864), but are not limited thereto.
  • the expression vector when using COS cells or NIH 3T 3 cells as host cells, the expression vector has the SV40 origin of replication, and is capable of autonomous growth in COS cells or NIH 3 T 3 cells. Further, those having a transcription promoter overnight, a transcription termination signal, and an RNA splice site can be used.
  • the expression vector is prepared by the DE AE-dextran method (Luthman, H. andd Magnusson, G. (1983) Nucleic Acids Res. 11, 1295-1308), calcium phosphate DNA co-precipitation method (Graham , FL and van der Eb, AJ (1973) Virol ogy 52, 456-457), and electric pulse drilling ('Neumann, E. et al.
  • the transformant obtained as described above can be cultured according to a conventional method, and the culture produces the target polypeptide inside or outside the cell.
  • Various commonly used cells can be appropriately selected depending on the host cell. If, in a medium such as RPMI 1 6 4 0 medium and Dulbecco's modified Eagle's medium (hereinafter referred to as "DME M"), can be used after the addition of serum components such as ⁇ shea calf serum as necessary.
  • DME M Dulbecco's modified Eagle's medium
  • the recombinant protein produced in the cells of the transformant or extracellularly by the above culture can be separated and purified by various known separation procedures utilizing the physical and chemical properties of the protein. it can.
  • Various types of liquid chromatography such as treatment with ordinary protein precipitants, ultrafiltration, molecular sieving chromatography (gel filtration), adsorption chromatography, ion exchange chromatography, affinity chromatography, high-performance liquid chromatography (HPLC) Dialysis, a combination thereof, and the like.
  • HPLC high-performance liquid chromatography
  • An example of an antibody that specifically binds to human oculospanin is a monoclonal antibody that specifically binds to human oculospanin, and the method of obtaining the antibody is as described below. .
  • myeloma preparation of myeloma cells
  • a method for producing a monoclonal antibody will be described in detail along the above steps, but the method for producing the antibody is not limited thereto.
  • antibody-producing cells other than spleen cells and myeloma may be used.
  • a Purification of antigen
  • human oculospanin prepared by the method described above or a part thereof can be used.
  • a membrane fraction prepared from the recombinant cells expressing human occulospanin, or the recombinant cells expressing human occulospanins themselves, and the protein of the present invention chemically synthesized using a method well known to those skilled in the art.
  • the partial peptide can also be used as an antigen.
  • the antigen obtained in step (a) is mixed with an adjuvant such as Freund's complete or incomplete adjuvant or potato pancreatic milk, and the animal is immunized as an immunogen.
  • an animal used for a known hybridoma production method can be used without any trouble.
  • a mouse, a rat, a goat, a sheep, a wedge, a horse, and the like can be used.
  • a mouse or rat as an animal to be immunized from the viewpoint of availability of myeloma cells to be fused with the isolated antibody-producing cells.
  • the strains of mice and rats actually used are not particularly limited.
  • mice for example, strains A, AKR, BALB / c, BDP, BA, CE, C3H-, 57BL, C57BR, C57L, DBA, FL, HTH, HT1, LP, NZB, NZW, RF, RIII, SJL, SWR, WB, 129, etc.
  • a rat for example, Low, Lewis, Spraque, Daeley, ACI, BN, Fischer, etc. can be used.
  • mice and rats can be obtained from experimental animal breeding distributors such as Japan Marie, Japan Charls River, Japan SLC and The Jackson Laboratories.
  • mice and the low strain in rats are particularly preferred as immunized animals in view of the compatibility with myeloma cells described below.
  • a mouse in which the biological mechanism for removing the self-progenitor is reduced that is, an autoimmune disease mouse, in consideration of the homology between the antigen and the human in the mouse.
  • the age at the time of immunization of these mice or rats is preferably 5 to 12 weeks, and more preferably 6 to 8 weeks.
  • a preferred method in the present invention is specifically described as follows, for example. That is, first, a membrane protein fraction, which is an antigen, or a cell that expresses the antigen is intradermally or intraperitoneally administered to an animal. However, in order to enhance the immunity efficiency, it is preferable to use both of them.
  • Intradermal administration is performed in the first half and intraperitoneal administration is performed only in the second half or the last time, whereby the immunity efficiency can be particularly increased.
  • the antigen administration schedule varies depending on the type of animal to be immunized, individual differences, and the like.In general, the antigen administration frequency is preferably 3 to 6 times and the administration interval is 2 to 6 weeks, and the administration frequency is 3 to 4 times and the administration interval. More preferably, 2 to 4 weeks. If the number of administrations is excessively increased, the antigen will be wasted, and if the administration interval is set too long, it is not preferable because the aging of the animals leads to a low activation of cells.
  • the dose of the antigen varies depending on the type of animal, individual differences, and the like, but is generally about 0.05 to 5 ml, preferably about 0.1 to 0.5 ml.
  • the booster immunization is performed 1 to 6 weeks, preferably 2 to 4 weeks, more preferably 2 to 3 weeks after the administration of the antigen as described above. If this booster is too late after 6 weeks or earlier than 1 week, the boost will have less effect.
  • the dose of the antigen for booster immunization varies depending on the type and size of the animal, but is generally 0.05 to 5 ml, preferably 0.1 to 0.
  • the volume should be about 5 ml, more preferably about 0.1 to 0.2 ml. Unnecessary large doses not only reduce the immune effect, but are not desirable for the immunized animals.
  • One to ten days, preferably two to five days, and more preferably two to three days after the above booster, spleen cells or lymphocytes containing bovine production cells are aseptically removed from the immunized animal.
  • the antibody titer is measured at that time, and the efficiency of the subsequent operation can be increased by using an animal having a sufficiently high antibody titer as a source of the antibody-producing cells.
  • the antibody titer used here includes various known techniques such as the RIA method, the ELISA method, the fluorescent antibody method, and the passive hemagglutination method. The detection sensitivity, speed, accuracy, and operation From the viewpoint of the possibility of automation, the RIA method or the ELISA method is more preferable.
  • the antibody titer in the present invention can be measured, for example, by the following procedure according to the ELISA method.
  • the purified or partially purified antigen is adsorbed on a solid surface such as a 96-well plate for ELISA, and the solid surface on which the antigen is not adsorbed is exposed to a protein irrelevant to the antigen, for example, a serum albumin (hereinafter referred to as “serum albumin”).
  • serum albumin hereinafter referred to as “serum albumin”.
  • an antibody against a mouse antibody labeled with an enzyme is added as a second antibody, and the antibody is bound to the mouse antibody.
  • a substrate of the enzyme is added, and the change in absorbance due to color development based on the decomposition of the substrate is measured. Calculate the value.
  • Isolation of antibody-producing cells from these spleen cells or lymphocytes can be performed by a known method (eg, Kohler et al., Nature, 256, 495, 1975; Kohler et al., Eur. J. Immnol., 6 , 511, 1977; ilstein et al., Nature, 266, 550, 1977; Walsh, Nature, 266, 495, 1977).
  • a known method eg, Kohler et al., Nature, 256, 495, 1975; Kohler et al., Eur. J. Immnol., 6 , 511, 1977; ilstein et al., Nature, 266, 550, 1977; Walsh, Nature, 266, 495, 1977.
  • MEM Eagle's minimum essential medium
  • myeoma cells used for cell fusion there is no particular limitation on the myeoma cells used for cell fusion, and they can be appropriately selected from known cell lines and used.
  • HGPRT Hoxan thi ne-guanine phoshor ibos y 1 tran sf erase
  • the power of using is preferred.
  • X63-Ag8 (X63), NSl-Ag4 / l (NS1), P3X63-Ag8.Ul (P3U1), X63-Ag8.653 (X63.653), SP2 / 0 -Agl 4 (SP2 / 0), MPC11-45.6TG1.7 (45.6TG), F0, S149 / 5XX0, BU.1, etc., 210.RSY3.Ag.1.2.3 (Y3), etc.
  • HGPRT-deficient strains can be obtained, for example, from American Type Culture Collection (ATCC).
  • These cell lines are prepared in an appropriate medium, for example, an 8-azaguanine medium [RPMI-1640 medium supplemented with glutamine, 2-mercaptoethanol, gentamicin, and fetal calf serum (hereinafter referred to as “FCS”). Medium supplemented with 8—azaguanine], Iscove's Modified Dulbecco's Medium (hereinafter referred to as “IMDM”), or Dulbecco's Modified Eagle Medium (hereinafter “DMEM”). 3-4 days before cell fusion, including normal medium [eg, containing 10% FCS] Subcultured ASF 1 0 4 medium (Ajinomoto Co., Ltd.)], set aside 2 X 1 0 7 or more cell number day of fusion.
  • 8-azaguanine medium RPMI-1640 medium supplemented with glutamine, 2-mercaptoethanol, gentamicin, and fetal calf serum
  • FCS fetal calf serum
  • FCS fetal calf serum
  • the fusion of the antibody-producing cells with the Mie cell is performed by a known method (Weir.DM, Handbook of Experimental Immunology Vol.I.II.III., Blackwe 11 Scientific Publications, Oxford ( 1987), Rabat, EA and Mayer, MM, Experimental.Immunochemistry, Charles C Thomas Publisher Spigfield, Illinois (1964)).
  • Such methods include, for example, a chemical method of mixing antibody-producing cells and myeoma cells in a high-concentration polymer solution such as polyethylene dalicol, a physical method using electrical stimulation, and the like. be able to.
  • specific examples of the above chemical methods are as follows.
  • the polyethylene glycol solution having a molecular weight of 1500 to 6000, preferably 2,000 to 4,000 is preferably used at a temperature of 30 to 40 ° C, preferably 35 to 38 ° C.
  • the fang-body producing cells and the Mie cell are mixed for 1 to 10 minutes, preferably for 5 to 8 minutes.
  • the method for selecting the hybridoma obtained by the above-mentioned cell fusion is not particularly limited.
  • a HAT (hypoxanthine'aminobuterin-thymidine) selection method CKohler et al., Nature, 256, 495 (1975); Mi 1 s tein at al., Nature 266, 550 (1977)].
  • This method is effective when obtaining hybridomas using myeloma cells of an HGPRT-deficient strain that cannot survive aminobuterin.
  • culturing unfused cells and hybridomas in a HAT medium only hybridomas having aminobuterin resistance can be selectively left and grown.
  • methylcellulose method soft agarose method
  • limiting dilution method can be used as the method for closing the hybridoma [eg, Barbara, BM and Stanley, MS: Selected Methods in Cellular Immunology, WH] See Freeman and Company, San Francisco (1980).
  • This closing Methods include limiting dilution, which involves diluting the culture so that one hybridoma is contained in each well of the plate, soft agar, which collects colonies by culturing in a soft agar medium, and micromanipulation.
  • One method is to take out and culture one cell at a time, and the other is to use a cell saw to separate one cell.
  • the limiting dilution method is particularly preferable.
  • a microplate is inoculated with a feeder such as a rat fetal fibroblast cell line or a normal mouse spleen cell, thymocyte, or ascites cell.
  • a feeder such as a rat fetal fibroblast cell line or a normal mouse spleen cell, thymocyte, or ascites cell.
  • the hybridoma is diluted in the medium so that it becomes 0.2 to 0.5 cells / 0.2 ml, and the diluted hybridoma suspension is added to each well in a volume of 0.2 mL.
  • Propagate hybridoma clones by adding 1 ml each and continuing cultivation for about 2 weeks while replacing about 1/3 of the medium with a new medium at regular intervals (for example, every 3 days). be able to.
  • cloning by limiting dilution is repeated 2 to 4 times, and those with a stable antibody titer are selected as anti-human oculospanin monoclonal antibody-producing hybridoma strains.
  • One of the eighty dorma strains cloned in this way was named 03B8-2C9-4F3, and was deposited at the National Institute of Advanced Industrial Science and Technology, Patent Organism Depositary (Tsukuba East, Ibaraki Prefecture). Deposit number FE RM BP— 08627 on February 17, 2004 at 1-chome, No. 1, Central No. 6).
  • the antibody titer in the present invention can be measured, for example, by the following procedure according to the ELISA method.
  • a solid-phase surface such as a 96-well plate for ELISA, and solid antigen-adsorbed cells are not adsorbed.
  • BSA serum serum albumin
  • an antibody against the mouse antibody labeled with an enzyme is added as the second antibody to bind to the mouse antibody. Calculate the antibody titer. Such screening may be performed after the hybridoma has been cloned as described above, or may be performed before it.
  • the hybridoma obtained by the above method can be stored in liquid nitrogen or in a freezer at 80 ° C or lower in a frozen state.
  • hybridomas are cultured by changing the medium from HT medium to normal medium.
  • Large-scale culture is performed by rotary culture using large culture bottles or single spinner culture.
  • a monoclonal antibody that specifically binds to the protein of the present invention can be obtained.
  • a hybridoma into the peritoneal cavity of a mouse of the same strain (for example, the above-mentioned BAL B / c) or NuZNu mouse, and growing the hybridoma, the monoclonal antibody of the present invention can be produced in a large amount. Can be obtained. If administered intraperitoneally, 2, 6, 10, 10 or 14 in advance (3 to 7 days before) — tetramethylpen
  • mineral oils such as (2, 6, 10, 1-tetramethyl pen t adecane) (pristane) produces more ascites.
  • mineral oils such as (2, 6, 10, 1-tetramethyl pen t adecane) (pristane) produces more ascites.
  • previously injected with immunosuppressive agents into the abdominal cavity of a mouse and the High Priestess dormer and the same strain After inactivation of the T cells, 1 after 2 days 0 0 6 -1 0 seven High Priestess de one Ma 'clonal cell Suspended in serum-free medium
  • a monoclonal antibody having a concentration of about 100 times or more as compared with that in a culture solution can be obtained.
  • the monoclonal antibody obtained by the above method can be purified, for example, by a method described in Weir, DM: Handbook of Experimental Immunology Vol. I, II, III, Blackwel Scientific Publications, Oxford (1978). That is, there are ammonium sulfate salting out method, gel filtration method, ion exchange chromatography method, affinity chromatography method and the like. Of these methods, the ammonium sulfate salting out method is repeated 3 to 4 times, preferably 3 to 6 times to obtain a monoclonal antibody. Can be purified. However, this method results in a very low yield of the purified monoclonal antibody.
  • a highly purified monoclonal antibody can be obtained in a high yield.
  • the combination and sequence of the ammonium sulfate salting out method and other methods include: a) ammonium sulfate salting out method—ion exchange chromatography—one method—gel filtration method, b) ammonium sulfate salting out method—ion exchange chromatography—affinity chromatography C) Ammonium sulfate salting out method-gel filtration method-affinity chromatography method, etc., but in order to obtain a monoclonal antibody with high purity and high yield, the above-mentioned c) Combinations are particularly preferred.
  • a commercially available monoclonal antibody purification kit for example, MAbTrapG11 kit; manufactured by Pharmacia
  • the monoclonal antibody thus obtained has high antigen specificity for human oculospan.
  • the determination of the isotype and subclass of the thus obtained monoclonal antibody can be performed as follows. First, as an identification method,
  • Immunoglobulin G is a light polypeptide chain having a molecular weight of about 230,000 (hereinafter, referred to as “light chain”) and a heavy chain having a molecular weight of about 500,000. It is composed of two polypeptide chains (hereinafter referred to as “heavy chains”). Both the heavy and light chains have a repeating structure of amino acid sequence conserved regions consisting of about 110 residues, and these are the basic units of the three-dimensional structure of IgG (hereinafter referred to as “domains”). Is configured. The heavy and light chains are composed of four consecutive and two domains, respectively.
  • the amino-terminal domain has a greater variation in amino acid sequence between antibody molecules than other domains, and this domain has a variable domain (V domain). ").
  • V domain variable domain
  • the heavy and light chain V domains complementarily associate to form a variable region.
  • the remaining domains form a constant region as a whole.
  • the constant region has a sequence that is characteristic of each animal species.For example, the mouse IgG constant region is different from the human IgG constant region because the mouse IgG constant region is different from the human IgG constant region. Recognized as foreign by the immune system, resulting in a Human Anti Mouse Antibody (HAMA) response.
  • HAMA Human Anti Mouse Antibody
  • mouse antibodies cannot be administered repeatedly to humans.
  • it is necessary to modify the antibody molecule so as not to generate a HAM ⁇ response while maintaining the specificity of the antibody.
  • such a domain generally has a long cylindrical structure in which two layers of antiparallel ⁇ -sheets consisting of three to five] chains are stacked.
  • variable region three loops are assembled in each of the V domains of the heavy and light chains to form an antigen-binding site.
  • Each of these loops is a complementarity determining region
  • CDR (. com rement arity determining region: J3 ⁇ 4 It is called “CDR.”)
  • the amino acid sequence variation is the most remarkable.
  • the portion of the variable region other than the CDR generally has a role of maintaining the structure of the CDR, and is called a “framework”.
  • Kabat et al. Collected a large number of primary sequences of the variable regions of the heavy and light chains, and created a table in which each primary sequence was classified into CDRs and frameworks based on the conservation of the sequences. SEQUENCES OF IMMUNOLOGICAL INTEREST, 5th edition, NIH publication, No. 91-3242, EA Kabatt et al.).
  • each framework Acid sequences were divided into subgroups with common features. Furthermore, it was found that there was a corresponding framework between human and mouse.
  • a chimeric antibody was proposed in which the variable region of a mouse-derived antibody was combined with a human-derived constant region (see Pro atl. Acad. Sci. U. S.A. 81, 6851-6855, (1984)).
  • chimeric antibodies still contain many non-human amino acid residues and can induce a HAMA response, especially when administered for long periods of time (Begent et al. Br. J. Cancer, 62., 487). , (1990)).
  • an antibody derived from a non-human mammal having a CDR to be transplanted is defined as "dona-one”, and a human antibody to which the CDR is to be transplanted is defined as "ax-e-u-ichi”.
  • the present invention also complies with this definition. I will.
  • a point to be considered when performing the CDR transplantation method is to preserve the structure of the CDR as much as possible and to maintain the activity of the immunoglobulin molecule. To achieve this goal:. (i) Which subgroup should Axep select?
  • amino acid is in the immediate vicinity of one of the CDRs
  • amino acid has a side chain atom within about 3 A of the CDR in the three-dimensional immunoglobulin model and is capable of interacting with the antigen or with the CDR of the humanized antibody.
  • the DNA encoding the heavy chain or light chain of the human oculospanin monoclonal antibody of the present invention is prepared by preparing mRNA from hybridoma cells producing the anti-human oculospanin monoclonal antibody. Then, the mRNA is converted to cDNA with reverse transcriptase, and then the DNA encoding the heavy chain or light chain of the antibody is isolated.
  • a guanidine-thiosinate-hot-phenol method for the extraction of mRNA, a guanidine-thiosinate-hot-phenol method, a guanidine-thiosineto-guanidine-hydrochloric acid method or the like may be employed, but the guanidine-thiosinate-cesium chloride method is preferred.
  • a poly (A) + RNA purification carrier such as oligo (dT) cellulose or oligo (dT) latex beads. It can be carried out by a method or a method of directly purifying from a cell lysate using the carrier.
  • the method for preparing the total RNA is as follows: Alkali sucrose density gradient centrifugation [see Dougherty, WG and Hiebert, E. (1980) Viology 101, 466-474], guanidine thiosineto phenol method
  • the guanidine thiocyanate trifluorocesium method, the phenol SDS method, and the like can also be employed. It is suitable.
  • Using the poly (A) + RNA obtained as described above as type III synthesize a single-stranded cDNA by reverse transcriptase reaction, and then synthesize a double-stranded cDNA from the single-stranded cDNA.
  • This method includes the SI nuclease method [Efstratiadis,
  • the double-stranded cDNA obtained in this manner is incorporated into a cloning vector, the resulting recombinant vector is introduced into a microorganism such as Escherichia coli, and transformed, and tetracycline resistance or ampicillin resistance is used as an indicator. As a result, a transformant can be selected. Transformation of Escherichia coli is performed by the Hanahan method [see Hanahan, D. (1983) J. Mol. Biol. 166, 557-580], that is, to competent cells prepared in the presence of chloridium chloride, magnesium chloride or rubidium chloride. The method can be carried out by adding the recombinant DNA vector. When a plasmid is used as a vector, it is necessary to have the above drug resistance gene. Cloning vectors other than plasmids, such as lambda phage, can also be used.
  • sense strand and antisense strand oligonucleotide primers corresponding to a part of the amino acid sequence are synthesized, and these are combined to form a polymer primer.
  • Perform a ze-chain reaction [see Saiki, RK, et al. (1988) Science 239, 487-49] to encode the desired anti-human oculospanin antibody heavy or light chain subunit.
  • the DNA fragment is amplified.
  • cDNA synthesized by reverse transcriptase reaction from mRNA of a hybridoma producing an anti-human-oculospanin monoclonal antibody can be used.
  • D NA fragment thus prepared can either be incorporated into a commercial directly bra corner de vector using kit Bok etc., the fragments were labeled with 32 P, 35 S or Pio Chin like, it
  • the target clone can also be selected by performing colony hybridization or plaque hybridization using the probe.
  • each subunit may be obtained by using a well-known method such as electrophoresis or force column chromatography.
  • a preferred method is to isolate the N-terminal amino acid sequence of each of the subunits using an automatic protein sequencer (for example, PPSQ-10 manufactured by Shimadzu Corporation) or the like.
  • an automatic protein sequencer for example, PPSQ-10 manufactured by Shimadzu Corporation
  • a method for collecting cDNAs encoding the respective subunits of the anti-human oculosvanin monoclonal antibody protein can be obtained by a known method [Maniatis, T., et al.
  • the oligonucleotide corresponding to the amino acid sequence Reotide is synthesized (in this case, either a nucleotide sequence deduced with reference to codon usage or a plurality of nucleotide sequences combining possible nucleotide sequences can be used, and in the latter case, inosine is included.
  • the sequence of the DNA obtained in this manner can be determined by, for example, the chemical modification method of maxam-gilpart [see Maxam, AM and Gilbert, W. (1980) in "Methods in Enzymology” 65, 499-576] It can be carried out by a xynucleotide chain termination method [see Messing, J. and Vieira, J. (1982) Gene 19, 269-276] and the like. '
  • DNA nucleotide sequencing can be performed efficiently and safely. From the nucleotide sequence of the DNA of the present invention thus determined and the N-terminal amino acid sequence of each of the heavy and light chains, all amino acids of the heavy and light chains of the monoclonal antibody of the present invention were obtained. The sequence can be determined.
  • Each of the heavy and light chains of immoglopurin consists of a variable region and a constant region, and the variable region further comprises a complementarity-determining region (hereinafter referred to as “CDR”; each of the heavy chain and the light chain has three sites). And the framework regions adjacent to them (four at each of the heavy and light chains).
  • CDR complementarity-determining region
  • the amino acid sequence of the constant region is common between antibodies having the same immunoglobulin subclass regardless of the type of antigen.
  • the amino acid sequence of the variable region especially the CDR
  • studies comparing the amino acid sequence data of many antibodies show that the CDR position and the length of the framework sequence are It is known that antibody subunits that succumb to the same subgroup are almost similar [Rabat, EA, el al. (1991) in "Sequence of Proteins of Immunological Interest Vol. II": US Department of Health and Human Services].
  • the chain length of F RH that is, the framework region closest to the N-terminal side of the heavy chain may be usually shorter than (30 amino acids).
  • the framework region has a minimum of 18 amino acids. Examples are known [see Kabat et al., Supra].
  • the chain length of the N-terminal framework region of the heavy chain is set to 18 amino acids or more and 30 amino acids or less, as long as the function as an anti-human oculospanin antibody is not impaired.
  • the peptide having the same amino acid sequence as the CDR of the light chain or the heavy chain determined as described above, or a peptide having a continuous partial amino acid sequence therein, is subjected to an artificial modification operation to obtain the CDR.
  • an artificial modification operation By approaching the three-dimensional structure formed in an anti-human oculospanin antibody molecule, it is possible to impart binding activity to human oculospanin alone [see, for example, US Pat. No. 5,331,7.3].
  • modified peptides having the same amino acid sequence as CDR or a contiguous partial amino acid sequence therein are also encompassed in the molecules of the present invention.
  • the cassette mutation method [Toshimitsu Kishimoto, "Natural Chemistry Laboratory 2 Nucleic Acid III Recombinant DNA Technology” 242 -251].
  • Such various types of DNA can be prepared by a conventional method such as the phosphophytate-triester method (see Hunkapiller, M., et al. (1984) Nature 310, 105-111). It can also be produced by chemical synthesis of nucleic acids.
  • the codon for the desired amino acid is known per se and may be arbitrarily selected. For example, the codon for the desired amino acid can be determined according to a conventional method in consideration of the codon usage of the host to be used.
  • nucleotide sequence codons can be partially modified by site-directed mutagenesis using a primer consisting of a synthetic oligonucleotide encoding the desired modification (site specific mutagenesis) according to a conventional method. Genesis) [Mark, DF, et al. (1984) Proc. Natl. Acad. Sci. USA 81, 5662-5666]. Whether or not a certain DNA hybridizes with DNA encoding the heavy chain or light chain of the anti-human occulospanin monoclonal antibody of the present invention can be determined by, for example, determining the DNA by a random primer method [ Feinberg, AP and Vogelstein, B. (1983) Anal, biochem.
  • the DNA to be examined is adsorbed on, for example, a nitrocellulose membrane or a nitrocellulose membrane and, if necessary, subjected to a treatment such as alkali denaturation, and then solidified by heating or ultraviolet rays.
  • This membrane contains 6 XSSC (1 XSSC is 0.15 M sodium chloride, 0.05 M trisodium citrate solution), 5% denhardt solution, 0.1% sodium dodecyl sulfate (SDS) immersed in Purehaipuri Daizeshiyo down solution from the incubated over 4 hours 5 5 ° C, the same pre-hybridization Dizeshiyo down solution pro portion of the previously prepared final specific activity 1 XI 0 6 cp mZm 1 and ⁇ so that the In addition, incubate at 60 ° C. Thereafter, the operation of washing the membrane with 6 XSSC at room temperature for 5 minutes is repeated several times, and further washing with 2 XSSC for 20 minutes, and then performing autoradiography.
  • a DNA encoding the heavy chain or light chain of the humanized anti-human oculospanin antibody of the present invention is hybridized from an arbitrary cDNA library or genomic library. Can be isolated [see Maniatis, T., et al. (1982) in Molecular Cloning A laboratory Manual Cold Spring Harbor Laboratory, NY.].
  • each of the DNAs obtained as described above into an expression vector, it is possible to introduce them into prokaryotic or eukaryotic host cells and to express each gene in those host cells.
  • the expression can be carried out by the same method as described in the section “(1) Preparation of antigen” in the section “5. Production of bovine human oculospanin antibody”.
  • the fraction containing the anti-human oculospanin antibody protein which is produced intracellularly or extracellularly in the transformant, can be obtained by various known protein separation procedures utilizing the physical and chemical properties of the protein. , Can be separated and purified. Examples of such a method include, for example, treatment with a normal protein precipitant, ultrafiltration, molecular sieve chromatography (gel filtration), adsorption chromatography, ion-exchange chromatography, affinity, and the like. Various chromatographies such as monochromatography, high performance liquid chromatography (HPLC), dialysis methods, and combinations thereof can be employed.
  • HPLC high performance liquid chromatography
  • the amino acids of the variable region should be changed so that the entire CDR sequence and some amino acid residues of the FR sequence are transferred to the human antibody. You need to design the array. This design follows the following method.
  • molecular modeling J the operation of predicting the tertiary structure from the primary sequence of an antibody molecule (hereinafter, this operation is referred to as “molecular modeling J”) has a limited accuracy of prediction, and amino acids that rarely appear in the subgroup to which the donor belongs are included. It is generally difficult to determine which amino acid residue, donor or acceptor, should be selected at such a position according to the method of Queen et al. According to, the chances of making such a decision can be significantly reduced.
  • the present inventors have provided a novel method for identifying amino acids derived from FRs of donna that are important for maintaining the structure and function of the donna CDR. Has improved.
  • a method for selecting an amino acid residue to be transplanted is as described below. If the amino acid sequences of the donor and the acceptor are aligned, and the amino acid residue is different at the corresponding position in the FRs, it is necessary to determine which residue to select, but this selection However, selection must be made so as not to impair the three-dimensional structure of the donor-derived CDR.
  • amino acid has a side chain atom within about 3 A of the CDR in a three-dimensional immunoglobulin model and is capable of interacting with the antigen or with the CDR of the humanized antibody.
  • the residue shown in 2) often exhibits the property of 3), and therefore, in the present invention, the requirement of 2) is deleted, and two new requirements are separately provided. That is, in the present invention, for amino acid residues on the donor FR to be transplanted with the CDR: a) The amino acid in the FR of the acceptor is rare at that position and the corresponding amino acid of Donna is Normal in position;
  • the amino acid residue is transplanted from the donor FR.
  • amino acids that are found at a frequency of 90% or more at the relevant position for antibodies of the same subclass are ⁇ normal, '' and amino acids that are found at a frequency of less than 10% are ⁇ rare.
  • the distance between two atoms is 0.5 A as the sum of each of the Van der Waals radii. If it is shorter than the value obtained by adding, it is estimated that the two atoms are in van der Waals contact.
  • the distance between polar atoms such as amide nitrogen and carbonyl oxygen in the main chain and side chain is shorter than the average hydrogen bond distance of 2.9 A plus 0.5 A, it is considered that hydrogen bonds exist between them. Estimated.
  • the distance between atoms with opposite charges was shorter than 2.85 A plus 0.5 A, it was assumed that an ion pair was formed between them.
  • Positions 4 103 are specified (all numbers represent amino acid numbers as defined in Kabat et al., Supra; the same applies hereinafter).
  • amino acid residues at these positions will be in contact with the amino acid residues of the CDR in two-thirds of the known antibody variable regions.
  • b) of "the amino acid is expected to interact with the antigen or CDR loop to be transplanted in the three-dimensional structural model of the amino acid in the three-dimensional structural model" satisfies the following requirements. means.
  • the position constitutes the contact surface between the heavy chain and the light chain means the following requirements. From the results of X-ray crystallography of the variable regions of various antibodies, it was found that in the light chain, the amino acid residues at positions 36, 38, 43, 44, 46, 49, 87, 98, heavy In the chain, the amino acid residues at positions 37, 39, 45, 47, 91, 103 and 104 were found to frequently make heavy-light chain contacts. Has been. In molecular modeling, the possibility of heavy chain-light chain contact is foreseen, and if that position matches any of the positions described above, transplantation of donor amino acid residues is prioritized. Otherwise, this requirement d) is not considered.
  • the DNA encoding the variable regions of the heavy and light chains of the humanized anti-human oculospanin antibody of the present invention can be produced by the method described below.
  • a plurality of polynucleotide fragments of 60 to 70 nucleotides consisting of the partial nucleotide sequence of the DNA are chemically synthesized so as to be alternate on the sense side and the antisense side, and then each polynucleotide fragment is synthesized.
  • the DNA is ligated and bound with DNA ligase to obtain a DNA having DNAs encoding the variable regions of the heavy chain and light chain of the desired humanized anti-human oculos vanin antibody.
  • DNA encoding the entire amino acid sequence of the variable region of Axepui can be separated from the human immunoglobulin and the CDR-encoding region can be subjected to nucleotide substitution by methods well known to those skilled in the art. Introduce an enzyme cleavage sequence. After cleaving the region with the corresponding restriction enzyme, a nucleotide sequence encoding the CDR of the donor is synthesized, and linked by DNA ligase to obtain the desired humanized anti-human urospanin antibody. DNA encoding the variable regions of the chains and light chains can be obtained. '
  • the desired humanized anti-human antibody is preferably prepared according to the following method (see Holton et al., Gene, 77, 61-68, (1989)). DNAs encoding the variable regions of the heavy and light chains of the oculospanin antibody can be obtained.
  • (A) and (B) two kinds of DNAs respectively encoding two kinds of amino acid sequences desired to be connected are referred to as (A) and (B) for convenience.
  • the primer is (D).
  • a chimera-type sense primer in which 20 to 30 nucleotides on the 3 ′ side of (A) and 20 to 30 nucleotides on the 5 ′ side of (B) are linked (hereinafter, this primer is referred to as (E) And an antisense primer complementary thereto (hereinafter, this primer is referred to as (F)).
  • PCR is performed using the sense primer (C) and the chimeric antisense primer (F) with the substrate as a substrate, whereby the 3 ′ end of (A) is 20 to 30 at the 5 ′ end of (B). Nucleotide-added DNA can be obtained (this newly obtained DNA is referred to as (G)).
  • the appropriate vector DNA containing (B) as a substrate and performing PCR using the antisense primer (D) and the chimeric sense primer (E) the 5 ′ of (B) can be obtained. DNA having 20 to 30 nucleotides at the 3 ′ terminal side of (A) added to the terminal can be obtained (this newly obtained DNA is referred to as (I-I)).
  • the (G) and (H) thus obtained are complementary at the 40 to 60 nucleotides on the 3 ′ side of (G) and the 40 to 60 nucleotides at the 5 ′ side of (H). It has a nucleotide sequence.
  • the amplified (G) and (H) are mixed and subjected to PCR, (G) and (H) become single-stranded in the first denaturation reaction, and most of the DNA is removed in the subsequent annealing reaction. It returns, but for some DNAs it forms a heteroDNA duplex that anneals with complementary nucleotide sequence regions.
  • the protruding single-stranded portion is repaired, and a chimeric DNA in which (A) and (B) are linked (hereinafter, this DNA is referred to as (I)) can be obtained. Further, by using this (I) as a substrate and performing PCR using a sense primer (C) and an antisense primer (D), (I) can be amplified.
  • this (I) as a substrate and performing PCR using a sense primer (C) and an antisense primer (D)
  • (I) can be amplified.
  • DNAs encoding the CDR regions of the heavy and light chains of the anti-human and oculospanin mouse monoclonal antibodies and the FR regions of the human immunoglobulin IgG are encoded. Furthermore, the above-mentioned ligation reaction is carried out using DNA encoding the secretion signal of human immunoglobulin IgG as (A) and (B) in case 'pi' case, respectively. Can be.
  • the codon for the desired amino acid is known per se, and its selection may be arbitrary.
  • the codon can be determined according to a conventional method in consideration of the codon usage of the host to be used. Partial modification of these nucleotide sequence codons can be performed by a conventional method using site specific mutagenesis (Mark, DF, Marker) using a primer consisting of a synthetic oligonucleotide encoding the desired modification. Natl. Acad. Sci. USA 81, 5662-5666) and the like. Therefore, when chemically synthesizing each primer, by designing each primer in advance to introduce point mutations, the variable regions of the heavy chain and light chain of the desired anti-human-clospanin antibody can be co-located. You can get the DNA you want. .
  • prokaryotic or eukaryotic host cells By incorporating each of the thus obtained DNAs of the present invention into expression vectors, prokaryotic or eukaryotic host cells can be transformed. Furthermore, by introducing an appropriate promoter and a sequence involved in expression into these vectors, each gene can be expressed in each host cell.
  • a recombinant anti-human oculospanin antibody can be easily produced with high yield and high purity.
  • a complete human antibody means a human antibody having only the gene sequence of an antibody derived from human chromosome.
  • the complete human ochulospanin human antibody is obtained by a method using a human antibody-producing mouse having a human chromosome fragment containing the genes for the H and L chains of the human antibody (Tomizuka, K. et al., Nature Genet. ics, 16, p.133-143, 1997 .; Kuroiwa, Y.et.al., Nuc. Acids Res., 26, p.3447-3448, 1998 .; Yoshida, H.et.al., Animal Cell Technology: Basic and Applied Aspects vol.
  • Methods for confirming that the recombinant anti-human oculospanin antibody thus produced specifically binds to human oculospanin include, for example, a method of evaluating a bovine titer during mouse immunization. Similar ELISA methods are preferred.
  • anti-human oculospanin antibodies obtained by the method described in the section “5. Production of anti-human oculopanin antibody”, an antibody or human antibody that neutralizes the biological activity of human oculopaninin. An antibody that specifically damages cancer cells that express oculospanin can be obtained. These antibodies can be used as a medicine, especially as a therapeutic agent against cancer, because they inhibit the biological activity of human toxourospanin in vivo, that is, inhibit the canceration of cells.
  • the activity of neutralizing the biological activity of human oculospanin by an anti-human oculospanin antibody in vitro can be measured by, for example, the activity of suppressing cell carcinogenesis in a cell overexpressing human oculospanin.
  • the mouse fibroblast cell line NIH3T3, which overexpresses human oculospanin, is cultured, and anti-human oculospanin antibodies are added to the culture system at various concentrations to form focus, colony formation and The inhibitory activity against spheroid proliferation can be measured.
  • In vitro anti-human oculospanin antibody-induced cancer cell-inducing activity includes, for example, antibody-dependent cytotoxicity exhibited by anti-human oculospanin antibody against cells overexpressing human oculospanin; It can be measured by complement-dependent cytotoxic activity or complement-dependent cytotoxic activity.
  • 293 T cells overexpressing human oculomouth spanin are cultured, anti-human oculospanin antibodies are added to the culture system at various concentrations, and mouse spleen cells are added for an appropriate time. After the culture, the cell death induction ratio with respect to cells overexpressing human occulospanin can be measured.
  • the therapeutic effect of anti-human urokurospanin antibodies on cancer using experimental animals was determined by, for example, By administering the same anti-oculospanin antibody to a transgenic animal overexpressing, the change in cancer cells can be measured.
  • the thus obtained antibody that neutralizes the biological activity of human oculospanin or the antibody that specifically damages cancer cells that express human oculospanin is used as a medicament especially for the purpose of treating cancer. It is useful as a composition or as an antibody for immunological diagnosis of such a disease.
  • Suitable types of cancer include, but are not limited to, skin cancer and melanoma, which is a type of skin cancer.
  • the present invention also provides a pharmaceutical composition
  • a pharmaceutical composition comprising a therapeutically effective amount of an anti-human oculospanin antibody and a pharmaceutically acceptable diluent, carrier, solubilizer, emulsifier, preservative and / or adjuvant.
  • the substance used in the formulation acceptable in the pharmaceutical composition of the present invention is preferably non-toxic to the person to whom the pharmaceutical composition is administered, preferably at the dosage and administration concentration.
  • the pharmaceutical composition of the present invention changes or maintains pH, osmotic pressure, viscosity, clarity, color, isotonicity, color, sterility, stability, dissolution rate, sustained release rate, absorption rate, and permeability. It can contain pharmaceutical substances for holding or holding. Pharmaceutical substances include, but are not limited to: glycine, alanine. Amino acids such as glutamine, asparagine, arginine or lysine, antimicrobial agents, ascorbic acid, sodium sulfate or bisulfite.
  • Antioxidants such as sodium phosphate, citrate, borate buffer, hydrogen carbonate, buffer such as tris-HCl solution, filler such as mannitol or glycine, ethylenediamine Chelating agents such as acetic acid (EDTA), caffeine, polyvinylpyrrolidine, complexing agents such as ⁇ -cyclodextrin / hydroxypropyl 1 / 3—cyclodextrin, and increasing amounts of glucose, mannose or dextrin; Ingredients, other carbohydrates such as monosaccharides, disaccharides and dulose, mannose dextrin, coloring , Flavoring agents, diluents, emulsifiers and hydrophilic polymers such as polyvinylpyrrolidine, low molecular weight polypeptides, salt-forming counterions, benzoconic chloride, benzoic acid, salicylic acid, thimerosal, phenethyl alcohol, methylparaben, propylparaben, Preservatives such as chlor
  • the amount of the substance for these preparations added is preferably 0.0'1 to 100 times, particularly preferably 0.1 to 10 times, the weight of the anti-human oculospanin antibody.
  • the composition of a suitable pharmaceutical composition in a formulation can be appropriately determined by those skilled in the art according to the disease to be applied, the administration route, and the like.
  • Excipients and carriers in pharmaceutical compositions may be liquid or solid. Suitable excipients and carriers may be water for injection, saline, artificial cerebrospinal fluid, or other substances commonly used for parenteral administration. Neutral saline or saline containing serum albumin can also be used as the carrier.
  • the pharmaceutical composition may also contain a Tris buffer with a pH of 7.0-8.5, an acetate buffer with a pH of 4.0-5.5, or sorpitol or other compounds.
  • the pharmaceutical composition of the present invention is prepared as an appropriate drug having the required purity in the selected composition and as a lyophilized product or liquid. Pharmaceutical compositions containing the human oculospanin antibodies can also be formed as lyophilized products using suitable excipients such as squeeze.
  • the pharmaceutical composition of the present invention can be prepared for parenteral administration or can be prepared for oral gastrointestinal absorption.
  • the composition and concentration of the preparation can be determined depending on the administration method, and the affinity of the anti-human oculospanin antibody for human oculospanin contained in the pharmaceutical composition of the present invention, that is, human. Based on these results, the higher the affinity (lower the Kd value), the lower the dose to humans, and the more effective it is for the dissociation constant (Kd value) for oculospanin.
  • the dose of the pharmaceutical composition of the present invention to a human can also be determined.
  • the dose should be about 0.:! ⁇ 100mkg may be administered once every 1-30 days.
  • Examples of the form of the pharmaceutical composition of the present invention include injections containing iv drops, suppositories, nasal drops, sublingual drops, and transdermal absorbents. 7. Search for directly interacting substances
  • Another embodiment of the present invention includes a drug design method based on the three-dimensional structure of the protein, which aims at obtaining a substance that suppresses the activity of human occulospanin.
  • a technique is known as a rational drug design method, and is used to search for a compound that efficiently inhibits or activates functions such as enzyme activity and binding to ligands, cofactors, or DNA. It's being used.
  • inhibitors of proteases which are already mentioned anti-HIV agents, are well known. It is considered that generally well-known techniques such as X-ray crystallography and nuclear magnetic resonance can be used in the three-dimensional structural analysis of the human oculospanin of the present invention.
  • a substance that suppresses the functions of human and oculospanins it is possible to design using a computer-drag design (CADD).
  • a computer-drag design is a low molecular weight compound (International Patent Application Publication No. WO99 / 58585) that inhibits the function of AP-1 which is expected as a new genomic drug for the treatment of rheumatoid arthritis. It has been done. In this way, substances that directly bind to human oculospanin or inhibit the function of human oculospanin by inhibiting the interaction of human oculospanin with other factors can be used. Obtainable.
  • Still another embodiment relates to a polypeptide to which the human oculospanin of the present invention associates, that is, a partner protein of human oculospanin. That is, the present invention relates to a method for screening a partner protein that regulates the activity of human oculospanin.
  • One embodiment of the screening method includes a step of contacting a test protein sample with human oculospanin and selecting a protein that binds to human oculospanin.
  • a method for example, there is a method of performing affinity purification of a protein binding thereto using purified human oculospanin.
  • An example of a specific method is as follows. A fusion of human oculospanin with a sequence consisting of six histidines as an affinity tag is prepared, and this is extracted with a cell extract (pre-charged to a nickel-agarose column).
  • the fractions that passed through the column were incubated at 4 ° C for 12 hours, and then the nickel-agarose carrier was added to the mixture and incubated at 4 ° C for 1 hour. After sufficiently washing the lug agarose carrier with a washing buffer, the protein in the cell extract that specifically binds to human oculospanin is eluted and purified by adding 10 OmM imidazole. , Determine this structure. In this way, a complex is formed with a protein that directly binds to human oculospanin and a protein that has no binding activity to human oculospanin but directly binds to human oculospanin as a subunit.
  • a functional class of a substance that inhibits the interaction between human oculospanin and the partner protein is obtained. It can be used for leaning.
  • a fusion protein of human oculospanin and daltathione S-transferase was prepared and bound to a microphone-mouth plate covered with an anti-glutathione S-transferase antibody. Thereafter, the biotinylated partner protein is brought into contact with the fusion protein, and binding to the fusion protein is detected with streptavidinated alkaline phosphatase.
  • a test substance is also added, and a substance that promotes or inhibits the binding between the fusion protein and the partner protein is selected.
  • a substance which directly acts on the fusion protein or a substance which directly acts on the partner protein is obtained. If the binding between the fusion protein and the partner protein is indirect and is via some other factor, for example, in the presence of a cell extract containing the factor, Atsushi is done. In this case, a substance that acts on the factor may be selected.
  • an anticancer agent such as a prostate can be prepared according to the test method using an expression vector of the human oculospanin gene described above.
  • Candidate substances useful as cancer therapeutics can be screened.
  • a polynucleotide having a nucleotide sequence encoding such a suppressor can be used as a gene for cancer. Can be used for treatment.
  • Such polynucleotides can be obtained, for example, by analyzing the amino acid sequence of the identified inhibitor, synthesizing an oligonucleotide probe consisting of a nucleotide sequence encoding the amino acid sequence, and synthesizing a cDNA library or genomic library. It can be obtained by performing cleaning.
  • the peptide having an inhibitory activity on the function of human oculospanin is derived from a randomly synthesized human epeptide library
  • a DNA comprising a nucleotide sequence encoding the amino acid sequence of the peptide is chemically synthesized. I do. '
  • a gene encoding the inhibitor thus obtained is incorporated into, for example, a viral vector, and the virus (detoxified) having the recombinant viral vector is transmitted to a patient.
  • Anticancer factors are produced in the patient's body and have the function of suppressing the growth of cancer cells, so that cancer can be treated.
  • Methods for introducing a gene therapy agent into cells include a gene transfer method using a viral vector or a non-viral gene transfer method (Nikkei Science, April 1994, 20-45). Page, Experimental Medicine Special Edition, 12 (15) (1994), Experimental Medicine Supplement “Basic Technology for Gene Therapy”, Yodosha (1996)) can be applied.
  • Non-viral gene transfer methods include direct injection of the expression plasmid into the muscle (DNA vaccine method), ribosome method, lipofectin method, and myelinomic method. Examples include a cloinjection method, a calcium phosphate method, and an electroreaction method, and a DNA vaccine method and a ribosome method are particularly preferable.
  • the gene therapy agent when administered by the in vivo method, it is administered by an appropriate administration route such as vein, artery, subcutaneous, intradermal, intramuscular, etc., depending on the disease, symptom and the like.
  • the gene therapy agent When administered by the in vivo method, the gene therapy agent is generally used as an injection or the like, but if necessary, a conventional carrier may be added.
  • ribosome preparations such as suspensions, freezers, and centrifugal concentrated freezers can be used.
  • a nucleotide sequence complementary to the nucleotide sequence shown in SEQ ID NO: 1 of the sequence listing or a nucleotide sequence complementary to a partial sequence of the sequence can be used for so-called antisense therapy.
  • the antisense molecule is a DNA consisting of usually 15 to 30 mer, or a phosphorothioate, methylphosphonate or morpholine thereof, which is complementary to a part of the nucleotide sequence shown in SEQ ID NO: 1 in the sequence listing.
  • stable RNA derivatives such as 2′-O-alkyl RNA.
  • antisense molecules are introduced into cells by methods well known in the art of the present invention, such as by microinjection, liposomal encapsulation, or expression using a vector having an antisense sequence. Can be.
  • Such an antisense therapy is described in SEQ ID NO:
  • a composition useful as a medicine containing the antisense oligonucleotide can be produced by a known method such as mixing a pharmaceutically acceptable carrier. Examples of such carriers and manufacturing methods are described in Applied Antisense Oligonucleotide Technology.
  • Antisense Ori Formulations containing tide may be mixed with excipients, diluents, etc. per se or as appropriate pharmacologically acceptable, and orally in tablets, capsules, granules, powders or syrups. Alternatively, it can be administered parenterally by injection, suppository, patch, or external preparation. These preparations may contain excipients (e.g., lactose, sucrose, dextrose, sugar derivatives such as mannitol, sorbitol; corn starch, potato starch, starch derivatives such as alpha starch, dextrin; crystalline cellulose).
  • excipients e.g., lactose, sucrose, dextrose, sugar derivatives such as mannitol, sorbitol; corn starch, potato starch, starch derivatives such as alpha starch, dextrin; crystalline cellulose.
  • Organic derivatives such as gum arabic; dextran; pullulan; and silicate derivatives such as light anhydrous silicic acid, synthetic aluminum silicate, calcium silicate, and magnesium aluminate silicate; hydrogen phosphate Phosphates such as calcium; carbonates such as calcium carbonate; inorganic excipients such as sulfates such as calcium sulfate; and lubricants (eg, stearic acid, stearic acid) Calcium stearate, such as calcium and magnesium stearate; talc Waxes such as beads, gays, etc .; boric acid; azidoic acid; sulfates such as sodium sulfate; glycosyl; fumaric acid; sodium benzoate; DL leucine; sodium lauryl sulfate, lauryl Lauryl sulfates such as magnesium sulfate; silicic acids such as silicic anhydride and silicic acid hydrate; and the above-mentioned starch derivatives
  • Colloidal clays such as beamum; metal hydroxides such as magnesium hydroxide and aluminum hydroxide; anionic surfactants such as sodium polyarylsulfate and calcium stearate; cations such as benzalkonium chloride.
  • Ionic surfactants; and nonionic surfactants such as polyoxyethylene alkyl ether, polyoxyethylene sorbin fatty acid ester, and sucrose fatty acid ester.
  • Stabilizer Paraoxybenzoic acid esters such as methylparaben and propylparaben; alcohols such as chlorobutanol, benzyl alcohol and phenylethyl alcohol; benzalkonium chloride; phenols such as phenol and cresol; thimerosal; And sorbic acid.
  • Flavoring agents for example, commonly used sweeteners, sour agents, flavors, etc.
  • additives such as diluents. And manufactured in a known manner.
  • a colloid dispersion system can be used in addition to the above.
  • the colloid dispersion system is expected to have the effect of increasing the stability of the compound in vivo and the effect of efficiently transporting the compound to a specific organ, tissue or cell.
  • Colloidal dispersions are not limited as long as they are commonly used, but are based on polymer complexes, nanocapsules, microspheres, peas, and oil-in-water emulsifiers, lipids including micelles, mixed micelles, and ribosomes. Ribosomes and vesicles of artificial membranes, which have the effect of efficiently transporting a compound to a specific organ, tissue or cell (Mannino et al. , Biotechniques, 1988, 6, 682; Blume and Cevc, Biochem. Et Biophys. Acta, 1990, 1029, 91; Lappalainen et al., Ant iviral Res., 1994, 23, 119; Chonn and Cul 1 is, Current Op. Biotech., 1995, 6, 698).
  • Monomembrane ribosomes can encapsulate a significant proportion of the aqueous buffer containing macromolecules, and the compound is encapsulated in this aqueous inner membrane, (Fraley et al., Trends ioiochem. Sci., 1981, 6, 77)
  • the composition of ribosomes is usually determined by lipids, especially phospholipids, especially phase transition temperatures. High lipid phospholipids are usually complexed with one or more steroids, especially cholesterol.
  • phosphatidyl compounds such as ngolipids, phosphatidylethanolamines, cereprosides and gangliosides, and particularly useful is diasylphosphatidylglycerol.
  • the lipid moiety contains 14-18 carbon atoms, especially 16-18 carbon atoms, and is saturated (double bond inside the 14-18 carbon atom chain).
  • the typical phospholipids are phosphatidylcholine, dipalmitoylphosphatidylcholine and distearoylphosphodies. And tidylcholine.
  • the targeting of colloidal dispersions can be either passive or active. Passive targeting is achieved by taking advantage of the ribosome's natural tendency to distribute to cells in the reticulum of organs containing sinusoids.
  • active targeting can be achieved, for example, by using a protein coat of a virus (Morish ita et al., Proc. Natl. Acad. Sci. (USA), 1993, 90,8474 :), a monoclonal antibody (or the like).
  • Binding specific ligands such as sugars, glycolipids or proteins (or suitable oligopeptide fragments thereof) to ribosomes, or organs and cells other than naturally occurring localized sites Techniques for modifying the ribosome by changing the composition of the ribosome to achieve distribution into types can be cited.
  • the surface of the targeted colloid dispersion can be modified in various ways.
  • lipid groups can be incorporated into the ribosome lipid bilayer to maintain the target ligand in tight association with the lipid bilayer.
  • Various linking groups can be used to link the lipid chain to the targeting ligand.
  • Target ligands that bind to specific cell surface molecules that are predominantly found on cells where delivery of the oligonucleotides of the invention are desired include, for example, (1) predominantly dependent on the cells where delivery is desired Specific to hormones, growth factors or their appropriate oligopeptide fragments, or (2) antigenic epitopes predominantly found on target cells, which bind to the specific cell receptor being expressed Or a suitable fragment thereof (eg, Fab; F (ab ′) 2), which binds to a polyclonal or monoclonal antibody.
  • the two or more bioactive agents can be combined and administered within a single liposome. Agents that increase the intracellular stability and Z or targeting of the contents can also be added to the colloid dispersion.
  • the dosage varies depending on symptoms, age, etc., but in the case of oral administration, the lower limit is 1 mg (preferably 30 mg) and the upper limit is 200 mg (preferably 150 mg) per dose. mg), in the case of injection, lower limit 0.1 lmg (preferably 5 mg), upper limit 100 mg (preferably 500 mg) subcutaneous injection, intramuscular injection Alternatively, it can be administered by intravenous injection.
  • lower limit 0.1 lmg preferably 5 mg
  • upper limit 100 mg (preferably 500 mg) subcutaneous injection, intramuscular injection Alternatively, it can be administered by intravenous injection.
  • each operation relating to gene manipulation is referred to as "Molecular Cloning" (written by Sambrook, J., Fritsch, EF and Maniatis, T., Cold Spring Harbor Laboratory Press, 1989). The method was used in accordance with the method described in (1), or when a commercially available reagent or kit was used, it was used in accordance with the instructions for the commercially available product.
  • Primer-1 is an oligonucleotide having 4 bases and a CACC added as a Kozak sequence upstream of the initiation codon of the human oculospanin gene, and has nucleotide numbers 1 to 23 of SEQ ID NO: 1 in the sequence listing.
  • CACC 4 nucleotide sequence
  • Primer 1-2 is an oligonucleotide consisting of a complementary strand of a nucleotide sequence consisting of nucleotide numbers 1043 to 1065 of SEQ ID NO: 1 in the sequence listing.
  • the PCR reaction was performed using PL ATINUM P fx DNA Polymerase (manufactured by Impitogen) according to the attached protocol. Specifically, to the obtained first strand cDNA dNA0. Lil, synthetic primer 1 and synthetic primer 2 having a concentration of lO pmol Zl were added to 1.5 1 and 10 XP fx Am plifaction Buffer 5, respectively.
  • UV—Free G DNA was purified from agarose gel by using el Purification Kit (manufactured by Invitrogen) according to the protocol attached thereto.
  • concentration of the purified cDNA was determined using lkb DNA L adder (Invitrogen, Inc.) using 1 PI Image Analysis Software Version 3.5 (Kodak Digital Science 'EDA S290: manufactured by Kodak). Was used as the concentration standard for the measurement.
  • the NM— 0 3 1 9 4 5c DNA obtained in Example 2 a) was converted into p ENTR / D _T ⁇ It was cloned into a PO vector.
  • the NM__ 0 3 1 9 4 5c DNA was mixed with the p ENTR ZD-TO PO vector to which the Topoisom erase was bound in the reaction buffer provided with the kit, and incubated at room temperature for 30 minutes.
  • Escherichia coli One S hot TO P10 Chemical 1y Chemical E.co1i (manufactured by Invitrogen) was transformed, and LB containing 50 Ig / m1 kanamycin was transformed. The cells were cultured on an agar medium. As a result, E. coli colonies that had grown with resistance to namycin were selected and cultured overnight at 37 in 1 ml of liquid TB medium containing 50 ng / m1 of kanamycin. the plasmid DNA was isolated and purified by using the M iniprep 9 6 K it (manufactured by Mi Ripoa Co.).
  • the gene was transferred to the expression vector PcDNA3.1 / DEST40 (Invitrogen) using the GAT EWAY TM system.
  • GAT EWAY TM LRC 1 onase TM Enzyme Mix manufactured by Invitrogen
  • 4 a LRR eaction Buffer 41 p ENTR / D -TO P O-NM_0 3 1 9 4 5 0.3 ⁇ g
  • Plasmid DNA (pcDNA3.1-DEST40-NM — 031945) was isolated and purified by using 1A GEN.
  • Plasmid pcDNA3.DEST40-NM-031945 obtained in Example 2 was transfected into NIH 3T3 cells as follows.
  • NIH 3 T 3 cells Transfection was carried out by lipofection using Lipofectamine TM 2000 Reagent from Invitrogen. That is, first, NIH 3T3 cells were grown in a 6-well plate until they became semi-confluent. Cells were washed once with DMEM containing no antibiotics and containing 10% fetal serum, and then D ⁇ ⁇ containing 100% antibiotics and containing 10% fetal serum. ⁇ was added.
  • DMEM serum-free medium
  • 2 ⁇ g of plasmid DNA pcDNA3.1-DEST40-NM_031945
  • DMEM serum-free medium
  • 4 ⁇ l of Lipofect amine TM 2000 Reagent were added and mixed.
  • the DNA solution and the Lipofect amine solution were mixed and left at room temperature for 20 minutes. Thereafter, a DNA-Lipofect amine mixed solution was added to the cells, and the cells were cultured at 37 ° C under 5% CO 2 .
  • DMEM 1 ml containing 1 0% ⁇ shea fetal serum after 4 hours the cells, 3 7 ° (:, and cultured overnight at 5% C 0 2 below.
  • NIH 3T3 cells obtained by transfection with cDNA-free negative control or pcDNA3.1-DEST40-NM-0331945 were washed with PBS (-) buffer (manufactured by Invitrogen). Cells were lysed in a sample buffer (manufactured by PIORAD) containing 2-mercaptoethanol for SDS-polyacrylamide electrophoresis (SDS-PAGE), and 12.5% polyacrylamide gel (e SDS-PAGE was carried out under reducing conditions using Pajiel E-T12.5 L AT-1 Co., Ltd.).
  • the PVD F membrane is blocked (once for 30 minutes at room temperature) with Block Ace (manufactured by Snow Brand Co., Ltd.), and then a plastic bag (trade name: Hyprivac, manufactured by Cosmo Bayo Co., Ltd.) ), Added anti-V5 antibody (1000-fold dilution) and 5 ml of block ess, and shaken at room temperature for 1 hour.
  • the membrane was taken out and washed with PBS containing 0.05% Tween 20 (hereinafter referred to as “0.05% Tween 20—PBS”) (1 minute at room temperature for 1 minute).
  • BAL B—3T3 cells American Type Culture Collection No. CCL-163
  • CS Dulbecco's modified Eagle medium containing 10% serum
  • DMEM Dulbecco's modified Eagle medium containing 10% serum
  • DNA solution and Geneporter TM 2 solution were mixed and left at room temperature for 20 minutes. Thereafter, DNA- G eneporter TM 2 mixture example pressurized to the cell (4m l / tray), 3 7 ° C, 5 % C 0 2 and cultured under.
  • the DMEM 5 0 m l / Torei added to cells containing 2 0% ⁇ shea serum after 4 hours, 3 7 "C, under 5% CO 2 and incubated overnight.
  • the cells cultured by the above method are washed with a PBS (-) buffer (manufactured by InV Itrogen). Collect the cells using a cell scraper (Sumitomo Bei-Cry 1, manufactured by K.K.) and suspend the cells in 8.0 mM ml of 5 mM Tris buffer. Leave the cell solution for 30 minutes at 4 ° C. Disrupt the cells with a D ounce Type B homogenizer (30 strokes). Centrifuge at 1000 G for 10 minutes and collect the supernatant. The supernatant is centrifuged at 800 G for 100 minutes with an ultracentrifuge (manufactured by Hitachi, Ltd.) to collect the precipitate. Enrich the membrane fraction with a sucrose density gradient.
  • mice To the precipitate, add 5 mM Tris buffer pH 8.050001, and homogenize the cell solution with a Dounce Tye B homogenizer (10 strokes).
  • the cell membrane fraction is identified by the western blotting method described in the section on expression confirmation and used as an immunogen.
  • Example 4 Immunization and cell fusion of mice
  • the spleen was excised from the mouse 3 days after the booster immunization, and the spleen was extracted with 20 mM HEPES buffer (pH 7.3), 350 mg / m 1 sodium bicarbonate, 0.05 mM ⁇ -mercaptoethanol, Serum-free RPMI containing 50 units / m 1 penicillin, 50 g / m 1 streptomycin, 300 / m 1 L-glutamic acid 1640 medium (10.4 gZ little RPMI 1 640 Nissi (1): Nissui Pharmaceutical Co., Ltd.) (hereinafter referred to as "serum-free RPMI medium") Place in 10 ml mesh (Celsto Reina: Falcon) Use spatula to crush. After the cell suspension that has passed through the mesh is centrifuged to precipitate spleen cells, the spleen cells are washed twice with serum-free RPMI medium, and then suspended in serum-free RPMI medium to determine the number of cells.
  • the following cell fusion procedure is performed while keeping the plastic centrifuge tube containing the pellets at 37 ° C in a beaker containing warm water. 1 ml of 50 (w / V) polyethylene glycol 150 (manufactured by Behringer Mannheim) was slowly added to the pellet while stirring the pellet at the tip of the pipette. Then, serum-free R pre-heated to 37 C Add 1 ml of PMI medium slowly in two portions, then add 7 ml of serum-free RPMI medium.
  • HAT medium J hypoxanthine 'aminopterin' thymidine medium
  • FCS hypoxanthine 'aminopterin' thymidine medium
  • thymus After extracting thymus from a 4- to 10-week-old BALB / c mouse female (purchased from Japan SLC), crush it with a spatula on a mesh (Cell Strainer; Falcon) and pass through the mesh
  • the cells thus obtained are washed twice with a hypoxanthine / thymidine medium containing 10% FCS (hereinafter referred to as “HT medium”; Kirk's Mannheim).
  • HT medium hypoxanthine / thymidine medium containing 10% FCS
  • Thymic cells of one mouse were suspended in 30 ml of HT medium containing 10% FCS, and the suspension was used as a feeder cell solution.
  • the culture solution containing the fused cells obtained in the above (4-2) is diluted 10 to 100 times with a feeder cell solution depending on the cell density, and the density of the fused cells is 5 cells / m 1 Dilute serially with feeder one cell solution to give 1 cell / m1 and 0.5 cell / 'm1.
  • Each sample prepared in this manner is dispensed into a 96-well microplate for cell culture at a ratio of 1001 / well, and cultured at 37 under 5% CO 2 for 5 days.
  • the maintenance culture of human oculospanin-expressing cells was performed using RPMI 1640 (manufactured by Inpitrogen), 10 fetal bovine serum (manufactured by Moregate Biotech), 20 mM HEPES (manufactured by Sigma), and 55 i ⁇ 2-mercaptoethanol (manufactured by Invitrogen). ) in the added medium (culture area), and 37 ° C, 53 ⁇ 4! C0 2 under. Transfer the cells expressing human occulospanin in the logarithmic growth phase to a cell culture flask at 2 ⁇ 10 4 cellsZcm 2 and culture for 3 days.
  • the 96-well U-bottom plate was centrifuged with HITACHI himac CF8DL at 15000 rpm for 1 minute (centrifugation condition 2), and the supernatant was removed with 200 1 chip. After hitting the side of the 96-well U-bottom plate to suspend the cells expressing human occulospanin, adjust the concentration to 10 / zg / ml and 5 ⁇ g / mK 2.5 ⁇ g / ml in a medium cooled on ice. Add the hybridoma culture supernatant ⁇ 1 / well. Incubate the 96-well U-bottom plate every 15 minutes at 4 ° C for 1.5 hours while stirring with a plate mixer (Fujirebio).
  • the human occulospanin-expressing cells obtained in Example 3 were cultured and grown in RPMI 1640 medium containing 10% FCS at 37 ° C and 5% carbon dioxide. From 1 X 1 0 7 cells suspension prepared in cell Zm 1, the U-shaped bottom 9 6-well microplate (manufactured by Nunc) dispensed at 5 0 1 Z Ueru min, centrifuged (9 0 X g , 4 ° C, 10 minutes). The supernatant is removed, the culture supernatant of the fused cells cultured in the above (4-1-3) is added at 50 1 / well, and the mixture is stirred. The mixture is allowed to stand on ice for 1 hour, and then centrifuged (90%). X g, ⁇ 0 min) and remove the supernatant. 1 0 0 0
  • FITC Fluorescein-5-isothiocyanate
  • the cell solution is added by adding 501, and the cells are fixed by leaving still on ice for 10 minutes. After centrifugation (90 X g, 4.C, 10 minutes) to remove the supernatant, wash again with buffer for flow cytometry 100.1 / well, and pellet the pellet by flow cytometry. 1. Use the suspension in the 100 ⁇ l ⁇ -well solution for re-use as a sample for single-hole cytometry. The FITC fluorescence intensity of the cells in each sample is measured with a flow cytometer (Epics Elite; manufactured by Cole Yuichisha) (excitation wavelength: 488 nm, detection wavelength: 530 nm).
  • a flow cytometer Epics Elite; manufactured by Cole Yuichisha
  • the FITC fluorescence intensity of a clearly higher value (approximately 100 to 100) was obtained than that of the human oculospanin-expressing cells (FITC fluorescence intensity of approximately 0.3) to which no fusion cell culture supernatant was added.
  • the fused cells corresponding to the indicated samples are selected.
  • the mouse-mouse hybridoma produced in Example 4 was replaced with 10% FCS.
  • AS F medium 1 in liters containing, 3 7 ° C, and cultured under 5% carbon dioxide, and grown until IX 1 0 6 cells Zm l.
  • the culture was centrifuged (1000 rpm, 2 minutes), the supernatant was discarded, and the precipitated cells were washed once with serum-free ASF medium, and resuspended in 1 liter of serum-free ASF medium. Incubate at 7 ° (: 5% carbon dioxide for 48 hours.
  • Each eluate was placed in a test tube containing 1.125 ml of 1 M Tris-hydrochloric acid (pH 9.0), and immediately after completion of the elution, a centrifugal tube type ultrafilter (Centreprep 10; (Grace Japan Co., Ltd.) and centrifuged at 30000 ⁇ g at 4 ° C. for 2 hours. After removing the filtrate collected in the lower part of the filter, add 15 ml of PBS to the upper part, and repeat the operation of centrifuging again at 30000 xg and 4 ° C for 2 hours five times. However, perform the fifth centrifugation until the liquid volume in the upper part of the filter becomes 0.5 ml, and use the liquid remaining in the upper part of the filter as the anti-human oculospanin antibody sample. ,
  • Antibody-dependent cytotoxicity is measured as an indicator of biological activity.
  • RPMI 164 medium Invitrogen, hereinafter referred to as RPMI medium
  • 10% fetal serum Methicillin
  • Effecta cells are prepared as follows. After counting the J774A.1 cells (Dainippon Pharmaceutical) in the presence of 10 O ng / ml of macrophage colony-stimulating factor (Sigma) for 3 days by trypan blue staining, the RPM I in the culture area to prepare to 1 X 1 0 6 cells / m 1. Seed 100 l (lxl 0 5 cells) on the 96-well round bottom microplate and centrifuge the plate at 150 rpm for 5 minutes. Incubate for 4 hours. To the positive control well, add l% Trion X-100 instead of effecta cells to completely kill BATDA-labeled human oculospanin-expressing cells.
  • Cell death induction rate (%) (count of each test well—count of negative control / well) / (count of positive control / one count of negative control) x l O 0
  • the obtained plasmid DNA was subjected to a reaction using Big Dye Terminatorv 3.0 Cyclic Sequencing Rapid Reaction Kit according to the protocol attached thereto, and then ABIPRI SM 310 Nucleotide sequence analysis was performed by DNA Analyzer (Applied Biosystems), and the GenBank accession number (ACCESSI ON NO. NM—O pen of the nucleotide sequence shown in 031945) was used. It was confirmed that the cDNA having the Leading Frame (SEQ ID NO: 1 in the sequence listing) was integrated into the pENT RZD—TOPO vector.
  • Gene transfer was performed to 1 / DEST 40 (manufactured by Invitrogen) using the GATEWAY TM system.
  • GATEWAY TM LRC 1 onase TM Enzyme Mix (manufactured by Invitrogen) 4 a LRR eaction Buffer 41, p ENTR / D -TO P O-NM_0 3 1 9 4 5 0.3 tg
  • pc Prepare 40.3 g of DNA 3.1 / DEST with TE buffer to make 20 n 1 and react at 25 ° C for 1 hour. After the reaction, add 2 ⁇ 1 Proteinase K and react at 37 ° C for 10 minutes. The resulting reaction product was used to transform E.
  • the gene was transferred to the expression vector PEF / DEST 51 (manufactured by Invitrogen) using the GAT EW AY TM system.
  • Escherichia coli One S hot TOPI 0 Chemical 1 y Chemical E. coli (manufactured by Nichroshen) was transformed, and LB agar containing 50 g nom 1 of ampicillin was transformed. Cultured on medium. As a result, Escherichia coli colonies that had grown with resistance to ampicillin were selected and cultured at 37 ° C. in 100 ml of liquid LB medium containing 50 igZml of ampicillin. Plasmid DNA (pEF-DEST5 ⁇ —031945) was isolated and purified using IKit (manufactured by QIAGEN). '
  • BAL B-3 T3 cells (RIKEN clone A31) were prepared using a modified Dulbecco's modified Eagle's medium (hereinafter DMEMJ: SIGMA) containing 10% serum (GIBC0) (hereinafter, BSJ). ) for cell culture were placed 150MmDish (culture area 148c m 2; after incubation at 37, 5% CO2 until semiconfluent E cement in manufactured IWAKI Co., Ltd.) 330 sheets, the EF-DEST51- NM- 031945 BAL B—Transfected into 3T3 cells. Transfection to BALB-3 T3 cells was performed by Lipofexion using Geneporter TM 2 Transcription Reagent from Gene Therapy Systems.
  • DMEMJ Dulbecco's modified Eagle's medium
  • GIBC0 GIBC0
  • the cells were washed once with a serum-free medium (DMEM), and then 20 ml of a serum-free medium (DMEM) was added.
  • DMEM serum-free medium
  • a serum-free medium DMEM
  • Opti-MEM I serum-free medium
  • Geneporter TM 2 Reagent 841 was added and mixed. The DNA solution and the Geneporter TM 2 solution were mixed and left at room temperature for 20 minutes.
  • DNA-Geneporter TM 2 mixture was added to the cells (1 ml / dish), and the cells were cultured at 37 ° C and 5% C% 2 . 3 hours 10% ⁇ shea serum medium was changed to DMEM 20m 1 / dish containing post, 3 7:, and cultured overnight at 5% C0 2 below.
  • plasmid p EF—DEST51—NM—031945 was introduced into 2293T cells as follows. Introduction into 293 T cells was performed using LIPOFEC TAMINE 2000 reagent (Invitorogen). 293 T cells were seeded at a density of 2.5 ⁇ 10 5 Z 9.2 cm 2 and cultured overnight at 37 ° C. under 5% carbon dioxide. In a 5 ml polypropylene tube, mix lOIL LIPOFEC TAM INE 2000 reagent with 2501 OPTI—MEM IR educed—Serum Medium, Invitorogen) and mix at room temperature. The reaction was performed for 5 minutes.
  • the cells cultured by the above method were washed with PBS (-) buffer (Dainippon Pharmaceutical Co., Ltd.). The cells were collected using a cell scraper (manufactured by IWAKI), and suspended in 5 mM Tris buffer 7.4230 ml. The cell solution was left for 30 minutes at 4 ° C. D ounce T ye B homogenizer (50 strokes) The cells were crushed with. The mixture was centrifuged at 100 G for 10 minutes using a centrifuge (manufactured by KUB0TA), and the supernatant was collected.
  • the supernatant was centrifuged for 800 minutes using an ultracentrifuge (BECKMAN), and the precipitate was collected.
  • 57% sucrose in Tris Buffer was added to 14 m, layered, and centrifuged at 78000 G for 16 hours at 4 ° C with a sucrose density gradient to collect the upper layer membrane fraction.
  • 55 ml of 5 mM Tris buffer (pH 7.4) was added to the membrane fraction, and the mixture was centrifuged at 78000 G for 60 minutes at 4 ° C to collect a precipitate.
  • 5 mM Tris buffer pH 7.4 15001 was added, and the cell solution was homogenized with a DounceTypeB homogenizer (10 strokes).
  • the cell membrane fraction was identified by the Wesin blotting method described in the section on expression confirmation.
  • mice [Example 8] Immunization and cell fusion of mice
  • the X10 7 cell expressing human occulospanin gene obtained in Example 7 was intraperitoneally administered to a 5-week-old female BAL BZc mouse (purchased from Nippon S.L.C.). Two, four, six, and eight weeks later, the same human and oculospanin gene-expressing cells (1 ⁇ 10 7 cells / mouse) were intraperitoneally administered for booster immunization.
  • the spleen was excised from the mice 4 days after the booster immunization, and this was supplemented with lOmM HE PES buffer (pI-I7.4), 0.02 mg / m1 sodium carbonate, 300 ⁇ g / ml L-daltamate.
  • Serum-free MEM medium (Eagle MEM medium “Nissi” (1): Nissui Pharmaceutical Co., Ltd., 9.4 g / L) (hereinafter referred to as “serum-free MEM medium J”)
  • the cell suspension was centrifuged to sediment the spleen cells, and the spleen cells were washed twice with serum-free MEM medium and suspended in serum-free MEM medium. After turbidity, the cell number was measured.
  • ⁇ medium a myeloma growth medium containing 15% FBS (GIBC0), 306 g / ml glutamic acid, and 0.05 mM ⁇ -mercaptoethanol (hereinafter referred to as “ ⁇ medium”) at 37 ° C. in the presence of 7% carbon dioxide gas in cell concentration and washed with the same serum-free MEM medium Mie port Ichima cells SP2 / 0 were cultured so as not to exceed 1 X 10 6 cells / m 1, were suspended in serum-free MEM medium measuring the number of cells did. An SP2 / 0 cell suspension equivalent to 1/5 spleen cells and a whole spleen cell suspension were mixed, centrifuged, and the supernatant was completely removed.
  • the following cell fusion operation was performed at room temperature in a plastic centrifuge tube containing a pellet.
  • lml of 40% (w / V) polyethylene glycol 4000 manufactured by Merck
  • 9 ml of the serum-free MEM medium was slowly added in three portions.
  • the tip of the pipet is adjusted to 2.5 ⁇ 10 6 cells / m 1 in hypoxanthine 'aminopterin' thymidine medium containing 20% FBS (hereinafter referred to as “HAT medium; SIGMA”).
  • HAT medium hypoxanthine 'aminopterin' thymidine medium containing 20% FBS
  • Dispense 10 On 1 Z-well into a 96-well microplate for cell culture and culture at 37 ° C (7% CO 2. One day later, add HAT medium 100/1 to all wells. The cells were exchanged every few days thereafter, and the thus obtained fused cells were subjected to screening by the limiting dilution method described below.
  • Each sample prepared in this manner was dispensed 100 1 / wel 1 each onto a 96weU plate in which 100 / LL 1 HY medium had been dispensed into each well in advance, and 37 days and 7% CO 2 gas for 10 days. Cultured.
  • Example 7 The cell membrane fraction obtained in Example 7 was dispensed at 50 ⁇ l / well at 1 g / ml into a 96-well EIA plate (C0STAR). After standing at 4 ° C for 1 day, shake the Array stock solution in the plate well, add 80 1 / wel 1 of 1% BSA to PBS (-), seal the plate, and seal at 4 ° until use. Saved in C. When used, the temperature was returned to room temperature, and the plate was washed three times with Serawasher (manufactured by Bio-Tec) through PBSCPBS-T containing 0.1% Tween20.
  • Serawasher manufactured by Bio-Tec
  • the absorbance at 405 nm and 630 nm was measured with a plate reader (Nalgenquie;> Tournal).
  • the HEK293 cultured cells obtained in Example 7 were grown and grown in a DMEM medium containing 10% FBS at 3.7 ° C. (5% CO 2 gas), and cultured for 24 hours after transfection. Dispense the cell suspension prepared in 2 ⁇ 10 7 cells 1 into 50-well 1-well cells in a 96-well microplate (Corning) and centrifuge
  • FITC Fluorescein-5-isothiocyanate
  • the FITC fluorescence intensity of the cells in each sample was measured by a flow cytometer (FC500; manufactured by BECKMAN) (excitation wavelength: 488 nm, detection wavelength: 530 nm).
  • FC500 flow cytometer
  • detection wavelength: 530 nm detection wavelength: 530 nm
  • Mouse one mouse High Priestess de one Ma prepared in Example 8 was suspended in HY medium so as to be lX10 6 cells / mI, and allowed to stand 37 ° C, 1% C0 2 3 days below.
  • the culture solution thus obtained was centrifuged (1600 rpm, 5 min), the supernatant was collected, and crude purification of IgG was performed under the following conditions.
  • Binding buffer PH7.0 (20mM Na 2 HP0 4 - 12H 2 0, 20mM Na 2 HP0 4 - 2H 2 0) elution buffer one: pH3.0 lOOmM glycine - HC1
  • ProteinG carrier (Amersiam Biosciences) was sampled in the required amount, ethanol was removed, washed once with ultrapure water, washed once with binding buffer, and then combined with binding buffer to prepare a 50% gel slurry. Protein G gel slurry was added to the supernatant, rotated overnight at 4 ° C, and then washed three times with binding buffer.After washing, an elution buffer was added to elute the antibody. The eluate was collected in a sample tube type ultrafilter (amicon Ultrafree-MC: Millipore, 5000 Xg, 4 ° C, 20 minutes) The eluate was added while removing the filtrate collected in the lower part of the filter so that the liquid volume in the upper part of the filter did not fall below 501. After all the eluate was added, PBS (-) was added. Add 3 times the volume of the eluate, exchange the buffer, and The solution remaining in the portion was used as an anti-anti-human oculospanin antibody sample.
  • Antibody-dependent cytotoxic activity was measured as an indicator of biological activity.
  • RPMI 164 medium manufactured by Invitrogen; hereinafter It was adjusted to 8 ⁇ 10 5 ce 11 s / 0.4 ml with RPMI medium.
  • Chromium—51 sodium chromate, manufactured by Amersham Biosciences
  • After addition of 401, 37 ° C, in the presence of 5% carbon dioxide Incubated for 2 hours.
  • Chromium-51-labeled human oculospanin-expressing cells were resuspended in 4 ml of RPMI medium, and purified mouse cells previously prepared at 5 ⁇ g / m 1 in RPMI medium.
  • 5-1 (1 ⁇ 10 4 ce 11 s) was seeded on a 96-well round bottom microplate to which oculospanin antibody 501 was added, and left at 4 for 30 minutes.
  • RPMI medium was added to the negative control well or background well in place of the purified mouse anti-human / oculospanin antibody. '
  • Effecta cells were prepared as follows. That BALB / c - collecting spleen cells according to a standard method from nu / nu mice (female, 7 weeks old), were prepared in 1 5 X 1 0 7 cells / 1 by the counting after RPMI medium with Toripanbu Le one staining.. Seed 10 ⁇ (1.5 x 10 6 eel 1 s) into the 96-well round bottom microplate and centrifuge the plate at 150 rpm for 5 minutes. Incubation was performed for 4 hours in the presence of 5% carbon dioxide. Positive control wells were supplemented with 2% Trion X-100 instead of effector cells to completely kill Chromium-51 labeled human oculospanin-expressing cells.
  • RPMI medium was added to the wells for background measurement instead of effector cells. After incubation for 4 hours, 501 culture supernatants were collected from each well and transferred to a 96-well luma plate (PerkinElmer). After drying at 50 ° C overnight, the amount of Chromium—51 in each well was measured using a microplate scintillation—Shion counter (Top Coat NTX, manufactured by PerkinElmer Inc.).
  • Cell death induction rate (%) (Count of each test well-count for background measurement / count of wells) / (Positive control-count of wells-for background measurement / force of wells) X 100
  • SEQ ID NO: 5 PCR sense primer for amplifying human occulospanin

Abstract

L'invention concerne un procédé pour détecter les cancers au moyen d'un oncogène, un procédé pour sélectionner des composés ayant des effets de prophylaxie et de traitement des cancers ainsi qu'une composition pharmaceutique pou la prophylaxie et le traitement des cancers. En particulier, l'invention concerne un procédé pour détecter les cancers au moyen de l'expression du gène humain oculospanin comme indice et une composition pharmaceutique comprenant un anticorps qui reconnaît spécifiquement l'oculospanin humain et possède une activité cytotoxique aux cellules d'un carcinome.
PCT/JP2004/003048 2003-03-10 2004-03-09 Anticorps contre un antigene specifique aux tumeurs WO2004081050A1 (fr)

Priority Applications (15)

Application Number Priority Date Filing Date Title
NZ542220A NZ542220A (en) 2003-03-10 2004-03-09 Antibody against the tumor specific antigen oculospanin
AT04718767T ATE434044T1 (de) 2003-03-10 2004-03-09 Antikörper gegen tumorspezifisches antigen als ziel
US10/548,688 US7855056B2 (en) 2003-03-10 2004-03-09 Antibody against tumor specific antigen as target
EP04718767A EP1602669B1 (fr) 2003-03-10 2004-03-09 Anticorps contre un antigène cible specifique de tumors
MXPA05009715A MXPA05009715A (es) 2003-03-10 2004-03-09 Anticuerpo contra antigeno especifico de tumor como objetivo.
DE602004021567T DE602004021567D1 (de) 2003-03-10 2004-03-09 Antikörper gegen tumorspezifisches antigen als ziel
DK04718767T DK1602669T3 (da) 2003-03-10 2004-03-09 Antistof mod et tumor-specifikt antigen som target
AU2004220182A AU2004220182B2 (en) 2003-03-10 2004-03-09 Antibody against tumor specific antigen as target
BRPI0408238-9A BRPI0408238A (pt) 2003-03-10 2004-03-09 anticorpo, método de detectar cáncer, conjunto para detecção de cáncer, e, composição farmacêutica para tratar cáncer
CA002518787A CA2518787A1 (fr) 2003-03-10 2004-03-09 Anticorps contre un antigene specifique aux tumeurs
US11/223,812 US7361340B2 (en) 2003-03-10 2005-09-09 Antibody against tumor specific antigen as target
NO20054631A NO20054631L (no) 2003-03-10 2005-10-07 Antistoff mot tumorspesifikt antigen som mal
HK06100346.5A HK1078593A1 (en) 2003-03-10 2006-01-09 Antibody against a tumor-specific antigen as target
US11/345,651 US20070025996A1 (en) 2003-03-10 2006-01-31 Oculospanin as a tumor specific antigen and methods and compositions utilizing same
US11/872,479 US7741447B2 (en) 2003-03-10 2007-10-15 Antibody against tumor specific antigen as target

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
JP2003063648 2003-03-10
JP2003-063648 2003-03-10

Related Child Applications (3)

Application Number Title Priority Date Filing Date
US10/548,688 A-371-Of-International US7855056B2 (en) 2003-03-10 2004-03-09 Antibody against tumor specific antigen as target
US11/223,812 Continuation US7361340B2 (en) 2003-03-10 2005-09-09 Antibody against tumor specific antigen as target
US11/345,651 Continuation-In-Part US20070025996A1 (en) 2003-03-10 2006-01-31 Oculospanin as a tumor specific antigen and methods and compositions utilizing same

Publications (1)

Publication Number Publication Date
WO2004081050A1 true WO2004081050A1 (fr) 2004-09-23

Family

ID=32984436

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2004/003048 WO2004081050A1 (fr) 2003-03-10 2004-03-09 Anticorps contre un antigene specifique aux tumeurs

Country Status (20)

Country Link
US (4) US7855056B2 (fr)
EP (1) EP1602669B1 (fr)
JP (1) JP4462964B2 (fr)
KR (1) KR20050116374A (fr)
CN (2) CN1784426A (fr)
AT (1) ATE434044T1 (fr)
AU (1) AU2004220182B2 (fr)
BR (1) BRPI0408238A (fr)
CA (1) CA2518787A1 (fr)
DE (1) DE602004021567D1 (fr)
DK (1) DK1602669T3 (fr)
ES (1) ES2327408T3 (fr)
HK (1) HK1078593A1 (fr)
MX (1) MXPA05009715A (fr)
NO (1) NO20054631L (fr)
NZ (1) NZ542220A (fr)
RU (1) RU2345090C2 (fr)
TW (1) TW200508249A (fr)
WO (1) WO2004081050A1 (fr)
ZA (1) ZA200507298B (fr)

Families Citing this family (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ZA200510282B (en) * 2003-07-02 2007-03-28 Genentech Inc Compositions and methods for the diagnosis and treatment of tumor
US20110195402A1 (en) * 2010-02-10 2011-08-11 Selinfreund Richard H Systems and methods for detecting drug use
EP2613380B1 (fr) 2010-11-05 2015-03-18 LG Chem, Ltd. Batterie rechargeable à stabilité améliorée
EP2589609A1 (fr) * 2011-11-03 2013-05-08 Pierre Fabre Medicament Protéine se liant à un antigène et son utilisation pour l'adressage d'un produit pour le traitement du cancer
US20140224373A1 (en) * 2013-02-12 2014-08-14 MPS Enterprises, Inc. Lay-flat hose for oilfield hydraulic fracturing operations
EP3736294A3 (fr) * 2014-10-10 2021-02-17 Innate Pharma Blocage de cd73
US11752577B2 (en) * 2018-11-15 2023-09-12 Samsung Display Co., Ltd. Laser apparatus and substrate etching method using the same
US20210106525A1 (en) * 2019-10-11 2021-04-15 Massachusetts Institute Of Technology Formulations for gastrointestinal delivery of oligonucleotides

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPH08191692A (ja) * 1994-08-04 1996-07-30 Bristol Myers Squibb Co ヒトガンに有効な新規なbr96抗体異性体
WO2001009187A2 (fr) * 1999-07-29 2001-02-08 Medarex, Inc. Anticorps monoclonaux humains diriges contre her2/neu
WO2003025138A2 (fr) * 2001-09-17 2003-03-27 Protein Design Labs, Inc. Procedes de diagnostic du cancer, compositions et procedes de criblage de modulateurs du cancer

Family Cites Families (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5331573A (en) * 1990-12-14 1994-07-19 Balaji Vitukudi N Method of design of compounds that mimic conformational features of selected peptides
US5728821A (en) * 1994-08-04 1998-03-17 Bristol-Myers Squibb Company Mutant BR96 antibodies reactive with human carcinomas
WO1999058515A1 (fr) 1998-05-08 1999-11-18 Toyama Chemical Co., Ltd. Nouveaux composes spiro et sels de ceux-ci, prophylaxie et medicaments contre des maladies auto-immunes et inhibiteurs de ap-1 contenant lesdits composes
US7442776B2 (en) * 1999-10-08 2008-10-28 Young David S F Cancerous disease modifying antibodies
US20020172986A1 (en) * 2000-08-21 2002-11-21 Leiby Kevin R. 23228, a novel human tetraspanin family member and uses thereof
US7361343B2 (en) * 2003-01-21 2008-04-22 Arius Research Inc. Cytotoxicity mediation of cells evidencing surface expression of CD63
US20030124579A1 (en) * 2001-09-05 2003-07-03 Eos Biotechnology, Inc. Methods of diagnosis of ovarian cancer, compositions and methods of screening for modulators of ovarian cancer
US20070042360A1 (en) * 2001-09-17 2007-02-22 Eos Biotechnology, Inc. Methods of diagnosis of cancer, compositions and methods of screening for modulators of cancer
KR20040101502A (ko) * 2002-04-16 2004-12-02 제넨테크, 인크. 종양의 진단 및 치료 방법 및 이를 위한 조성물
US7393531B2 (en) * 2003-01-21 2008-07-01 Arius Research Inc. Cytotoxicity mediation of cells evidencing surface expression of MCSP
US7361342B2 (en) * 2003-01-21 2008-04-22 Arius Research Inc. Cancerous disease modifying antibodies
ZA200510282B (en) * 2003-07-02 2007-03-28 Genentech Inc Compositions and methods for the diagnosis and treatment of tumor

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPH08191692A (ja) * 1994-08-04 1996-07-30 Bristol Myers Squibb Co ヒトガンに有効な新規なbr96抗体異性体
WO2001009187A2 (fr) * 1999-07-29 2001-02-08 Medarex, Inc. Anticorps monoclonaux humains diriges contre her2/neu
WO2003025138A2 (fr) * 2001-09-17 2003-03-27 Protein Design Labs, Inc. Procedes de diagnostic du cancer, compositions et procedes de criblage de modulateurs du cancer

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
AZORSA D.O. ET AL: "A general approach to the generation of monoclonal antibodies against members of the tetraspanin superfamily using recombinant GST fusion proteins", J. IMMUNOL. METHODS, vol. 229, no. 1-2, 1999, pages 35 - 48, XP002194216 *
LONGO N. ET AL: "Regulatory role of tetraspanin CD9 in tumor-endothelial cell interaction during transendothelial invasion of melanoma cells", BLOOD, vol. 98, no. 13, 2001, pages 3717 - 3726, XP002290253 *
MAMMALIAN GENE COLLECTION PROGRAM TEAM: "Generation and initial analysis of more than 15,000 full-length human and mouse cDNA sequences", PNAS USA, vol. 99, no. 26, 2002, pages 16899 - 16903, XP002964739 *
WISTOW G. ET AL: "Expressed sequence tag analysis of human RPE/choroid for the NEIBank Project: Over 6000 non-redundant transcripts, novel genes and splice variants", MOL. VIS., vol. 8, 2002, pages 205 - 220, XP002979786 *

Also Published As

Publication number Publication date
US20070025996A1 (en) 2007-02-01
ES2327408T3 (es) 2009-10-29
DE602004021567D1 (de) 2009-07-30
RU2345090C2 (ru) 2009-01-27
JP2004290187A (ja) 2004-10-21
US7741447B2 (en) 2010-06-22
EP1602669A4 (fr) 2006-08-09
AU2004220182A1 (en) 2004-09-23
CN102151334A (zh) 2011-08-17
EP1602669B1 (fr) 2009-06-17
HK1078593A1 (en) 2006-03-17
EP1602669A1 (fr) 2005-12-07
NZ542220A (en) 2008-04-30
US20080146785A1 (en) 2008-06-19
CA2518787A1 (fr) 2004-09-23
NO20054631D0 (no) 2005-10-07
ZA200507298B (en) 2006-06-28
NO20054631L (no) 2005-12-09
DK1602669T3 (da) 2009-09-14
TW200508249A (en) 2005-03-01
KR20050116374A (ko) 2005-12-12
US7855056B2 (en) 2010-12-21
AU2004220182B2 (en) 2007-12-06
MXPA05009715A (es) 2005-10-18
US20070166312A1 (en) 2007-07-19
JP4462964B2 (ja) 2010-05-12
US7361340B2 (en) 2008-04-22
BRPI0408238A (pt) 2006-03-01
US20060127405A1 (en) 2006-06-15
ATE434044T1 (de) 2009-07-15
CN1784426A (zh) 2006-06-07
RU2005128277A (ru) 2006-06-10

Similar Documents

Publication Publication Date Title
JP6440759B2 (ja) 破骨細胞関連蛋白質Siglec−15を標的とした抗体
KR101132579B1 (ko) 파골 세포 관련 단백질을 표적으로 한 항체
US9062100B2 (en) Anti-human TROP-2 antibody having anti-tumor activity in vivo
TWI443109B (zh) 抗epha2抗體
US7741447B2 (en) Antibody against tumor specific antigen as target
JP2011516078A (ja) 癌におけるegfr突然変異を検出する組成物および方法
WO2016171107A1 (fr) Détection de fgfr2
JP4633491B2 (ja) 破骨細胞関連タンパク質を標的とした抗体
TW201023893A (en) Antibodies recognizing oxygen-regulated protein 150 expressed on cancer cells and methods of using same
WO2022075482A1 (fr) Médicament pour le traitement du cancer
KR20140144934A (ko) Cthrc1의 발현 및 활성 억제제를 유효성분으로 포함하는 췌장암 치료 및 전이억제용 조성물
JP2004267118A (ja) 癌遺伝子及びその用途

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BW BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NA NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): BW GH GM KE LS MW MZ SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IT LU MC NL PL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

DPEN Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed from 20040101)
121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 1-2005-501632

Country of ref document: PH

WWE Wipo information: entry into national phase

Ref document number: 170641

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: 542220

Country of ref document: NZ

WWE Wipo information: entry into national phase

Ref document number: 2004718767

Country of ref document: EP

Ref document number: 2005/07298

Country of ref document: ZA

Ref document number: 2518787

Country of ref document: CA

Ref document number: 05091402

Country of ref document: CO

Ref document number: 1020057016909

Country of ref document: KR

Ref document number: 2005128277

Country of ref document: RU

Ref document number: 11223812

Country of ref document: US

Ref document number: 200507298

Country of ref document: ZA

WWE Wipo information: entry into national phase

Ref document number: PA/a/2005/009715

Country of ref document: MX

WWE Wipo information: entry into national phase

Ref document number: 01835/KOLNP/2005

Country of ref document: IN

Ref document number: 1835/KOLNP/2005

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: 2004220182

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 1200501478

Country of ref document: VN

ENP Entry into the national phase

Ref document number: 2004220182

Country of ref document: AU

Date of ref document: 20040309

Kind code of ref document: A

WWP Wipo information: published in national office

Ref document number: 2004220182

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 20048126663

Country of ref document: CN

WWP Wipo information: published in national office

Ref document number: 2004718767

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 1020057016909

Country of ref document: KR

ENP Entry into the national phase

Ref document number: PI0408238

Country of ref document: BR

NENP Non-entry into the national phase

Ref country code: JP

WWW Wipo information: withdrawn in national office

Ref document number: JP

WWP Wipo information: published in national office

Ref document number: 11223812

Country of ref document: US

WWE Wipo information: entry into national phase

Ref document number: 2007166312

Country of ref document: US

Ref document number: 10548688

Country of ref document: US

WWP Wipo information: published in national office

Ref document number: 10548688

Country of ref document: US